Language selection

Search

Patent 3133005 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 3133005
(54) English Title: COMBINATIONS OF RAD51 AND PARP INHIBITORS
(54) French Title: COMBINAISONS D'INHIBITEURS DE RAD51 ET DE PARP
Status: Report sent
Bibliographic Data
(51) International Patent Classification (IPC):
  • A61K 31/445 (2006.01)
  • A61K 31/4184 (2006.01)
  • A61K 31/4462 (2006.01)
  • A61K 31/4465 (2006.01)
  • A61K 31/496 (2006.01)
  • A61K 31/4985 (2006.01)
  • A61K 31/502 (2006.01)
  • A61K 31/55 (2006.01)
  • A61P 25/28 (2006.01)
  • A61P 35/00 (2006.01)
  • A61P 37/00 (2006.01)
(72) Inventors :
  • DAY, MELINDA (United States of America)
  • CASTRO, ALFREDO C. (United States of America)
  • MACLAY, TYLER (United States of America)
  • MCCOMAS, CASEY CAMERON (United States of America)
  • MILLS, KEVIN (United States of America)
  • VACCA, JOSEPH (United States of America)
(73) Owners :
  • CYTEIR THERAPEUTICS, INC. (United States of America)
(71) Applicants :
  • CYTEIR THERAPEUTICS, INC. (United States of America)
(74) Agent: SMART & BIGGAR LP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2020-03-25
(87) Open to Public Inspection: 2020-10-01
Examination requested: 2022-09-29
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2020/024601
(87) International Publication Number: WO2020/198298
(85) National Entry: 2021-09-08

(30) Application Priority Data:
Application No. Country/Territory Date
62/823,556 United States of America 2019-03-25

Abstracts

English Abstract

This application is directed to inhibitors of RAD51 represented by the following structural formula: in combination with a PARP inhibitor, and methods for their use, such as to treat cancer, autoimmune diseases, immune deficiencies, or neurodegenerative diseases.


French Abstract

Cette invention concerne des inhibiteurs de RAD51 représentés par la formule structurale suivante, en combinaison avec un inhibiteur de PARP et leurs procédés d'utilisation, tels que le traitement du cancer, de maladies auto-immunes, de déficiences immunitaires ou de maladies neurodégénératives.

Claims

Note: Claims are shown in the official language in which they were submitted.


CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
CLAIMS
1. A combination therapy comprising a compound represented by Formula (I):
, N
/ \ 0 R3
41k ,oS x5 y
6
R2
H N
Formula (I);
or a pharmaceutically acceptable salt thereof, wherein:
the thiazole ring is optionally substituted with ¨F or ¨C1;
Cy is -(C3¨C7)cycloalkyl, bridged (C6¨C12) cycloalkyl, or a 4-12 membered
heterocyclic ring, each of which is optionally substituted with one or more
groups selected
from the group consisting of halogen, -OH, (C1-C4)alkyl, and (C1-C4)alkoxy;
when X5 is connected with a nitrogen ring atom of Cy, X5 is absent;
when X5 is connected with a carbon ring atom of Cy, X5 is NRa or 0;
X6 is NRa or 0;
le is (Ci¨Cs)alkyl;
R3 is (Ci¨Cs)alkyl, -CH2-phenyl, -(C3¨C7)cycloalkyl, -CH2-(C3¨C7)cycloalkyl,
-CH2-monocyclic 3-7 membered heterocyclic ring, or monocyclic 3-7 membered
heterocyclic
ring, wherein the (Ci¨Cs)alkyl, -(C3¨C7)cycloalkyl, phenyl or monocyclic 3-7
membered
heterocyclic ring represented by R3 or in the group represented by R3 is
optionally substituted
with one or more groups selected from the group consisting of halogen, -OH,
(C1-C4)alkyl,
halomethyl, halomethoxy, -CN, and (C1-C4)alkoxy;
R2 is _NR
aC(0)0(C1-C4)alkyl; -NRaC(0)NRa(C1-C4)alkyl; -NRaC(0)0(C2-
C4)alkenyl;
-NRaC(0)NRa(C2-C4)a1keny1; -NRaC(0)0-(C3-C6)cyc1oa1ky1; -NRaC(0)NRa-(C3-
C7)cycloalkyl; -NRaC(0)0-pheny1; -NRaC(0)NRa-pheny1; -NRaC(0)0-monocyc1ic 3-7
membered heterocyclic ring; -NRaC(0)NRa-monocyc1ic 3-7 membered heterocyclic
ring; -
NRaC(0)0-monocyc1ic 5-6 membered heteroaromatic ring; -NRaC(0)NRa-monocyc1ic 5-
6
membered heteroaromatic ring;
wherein the (C1¨C4)alkyl and the (C2-C4)alkenyl in the group represented by R2
are
each optionally and independently substituted with one or more groups selected
from the
group consisting of halogen, N3, ¨0Ra, -NRaRa, -(C3¨C6)cycloalkyl, phenyl, a
monocyclic 3-
7 membered heterocyclic ring, and a monocyclic 5-6 membered heteroaromatic
ring;
106

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
wherein the (C3¨C7)cycloalkyl in the group represented by R2 is optionally
substituted
with one or more groups selected from the group consisting of halogen, -CH3,
=0, -0Ra and
_NRaRa;
wherein the phenyl in the group represented by R2 is optionally substituted
with one
or more groups selected from the group consisting of halogen, -CH3,
halomethyl,
halomethoxy,
-CN, -0Ra, and -N3;
wherein the heterocyclic ring in the group represented by R2 is optionally
substituted
with one or more groups selected from the group consisting of =0, halogen,
¨0Ra, -CH3,
halomethyl, and halomethoxy;
wherein the heteroaromatic ring in the group represented by R2 is optionally
substituted with one or more groups selected from the group consisting of
halogen, -CN, -
CH3, halomethyl, halomethoxy, -0Ra and -NRaRa; and
each Ra is independently ¨H or -CH3, and
a PARP inhibitor or a pharmaceutically acceptable salt thereof
2. The combination therapy of claim 1, wherein the PARP inhibitor is
olaparib.
3. The combination therapy of claim 1, wherein the compound of Formula (I)
has the
following structure:
0
I (--)" 'NH
0
/0
OAN SO
HNI
4. A pharmaceutical composition comprising a compound represented by
Formula (I):
\ 0 R3
R2 4410 ,Os X5Lxµ
HN \O
141
Formula (I);
or a pharmaceutically acceptable salt thereof, wherein:
107

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
the thiazole ring is optionally substituted with ¨F or ¨C1;
Cy is -(C3¨C7)cycloalkyl, bridged (C6¨C12) cycloalkyl, or a 4-12 membered
heterocyclic ring, each of which is optionally substituted with one or more
groups selected
from the group consisting of halogen, -OH, (C1-C4)alkyl, and (C1-C4)alkoxy;
when X5 is connected with a nitrogen ring atom of Cy, X5 is absent;
when X5 is connected with a carbon ring atom of Cy, X5 is NRa or 0;
X6 is NRa or 0;
le is (Ci¨Cs)alkyl;
R3 is (Ci¨Cs)alkyl, -CH2-phenyl, -(C3¨C7)cycloalkyl, -CH2-(C3¨C7)cycloalkyl,
-CH2-monocyclic 3-7 membered heterocyclic ring, or monocyclic 3-7 membered
heterocyclic
ring, wherein the (Ci¨Cs)alkyl, -(C3¨C7)cycloalkyl, phenyl or monocyclic 3-7
membered
heterocyclic ring represented by R3 or in the group represented by R3 is
optionally substituted
with one or more groups selected from the group consisting of halogen, -OH,
(C1-C4)alkyl,
halomethyl, halomethoxy, -CN, and (C1-C4)alkoxy;
R2 is _NR
aC(0)0(C1-C4)alkyl; -NRaC(0)NRa(C1-C4)alkyl; -NRaC(0)0(C2-
C4)alkenyl;
-NRaC(0)NRa(C2-C4)a1keny1; -NRaC(0)0-(C3-C6)cyc1oa1ky1; -NRaC(0)NRa-(C3-
C7)cycloalkyl; -NRaC(0)0-pheny1; -NRaC(0)NRa-pheny1; -NRaC(0)0-monocyc1ic 3-7
membered heterocyclic ring; -NRaC(0)NRa-monocyc1ic 3-7 membered heterocyclic
ring; -
NRaC(0)0-monocyc1ic 5-6 membered heteroaromatic ring; -NRaC(0)NRa-monocyc1ic 5-
6
membered heteroaromatic ring;
wherein the (C1¨C4)alkyl and the (C2-C4)alkenyl in the group represented by R2
are
each optionally and independently substituted with one or more groups selected
from the
group consisting of halogen, N3, ¨0Ra, -NRaRa, -(C3¨C6)cycloalkyl, phenyl, a
monocyclic 3-
7 membered heterocyclic ring, and a monocyclic 5-6 membered heteroaromatic
ring;
wherein the (C3¨C7)cycloalkyl in the group represented by R2 is optionally
substituted
with one or more groups selected from the group consisting of halogen, -CH3,
=0, -0Ra and
-NRaRa;
wherein the phenyl in the group represented by R2 is optionally substituted
with one
or more groups selected from the group consisting of halogen, -CH3,
halomethyl,
halomethoxy,
-CN, -0Ra, and -N3;
108

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
wherein the heterocyclic ring in the group represented by R2 is optionally
substituted
with one or more groups selected from the group consisting of =0, halogen,
¨0Ra, -CH3,
halomethyl, and halomethoxy;
wherein the heteroaromatic ring in the group represented by R2 is optionally
substituted with one or more groups selected from the group consisting of
halogen, -CN, -
CH3, halomethyl, halomethoxy, -0Ra and -NRaRa; and
each Ra is independently ¨H or -CH3, and
a PARP inhibitor or a pharmaceutically acceptable salt thereof
5. The pharmaceutical composition of claim 4, wherein the PARP inhibitor is
olaparib.
6. The pharmaceutical composition of claim 4, wherein the compound of
Formula (I)
has the following structure:
0
0
,0
OAN SO
HN
7. A kit comprising a compound represented by Formula (I):
\ 0 R3
R2 40 ,OS = X5L-)(
HN
Formula (I);
or a pharmaceutically acceptable salt thereof, wherein:
the thiazole ring is optionally substituted with ¨F or ¨C1;
Cy is -(C3¨C7)cycloalkyl, bridged (C6¨C12) cycloalkyl, or a 4-12 membered
heterocyclic ring, each of which is optionally substituted with one or more
groups selected
from the group consisting of halogen, -OH, (C1-C4)alkyl, and (C1-C4)alkoxy;
when X5 is connected with a nitrogen ring atom of Cy, X5 is absent;
when X5 is connected with a carbon ring atom of Cy, X5 is NRa or 0;
X6 is NRa or 0;
109

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
le is (C1¨05)alkyl;
R3 is (Ci¨05)alkyl, -CH2-phenyl, -(C3¨C7)cycloalkyl, -CH2-(C3¨C7)cycloalkyl,
-CH2-monocyclic 3-7 membered heterocyclic ring, or monocyclic 3-7 membered
heterocyclic
ring, wherein the (Ci¨05)alkyl, -(C3¨C7)cycloalkyl, phenyl or monocyclic 3-7
membered
heterocyclic ring represented by R3 or in the group represented by R3 is
optionally substituted
with one or more groups selected from the group consisting of halogen, -OH,
(Ci-C4)alkyl,
halomethyl, halomethoxy, -CN, and (Ci-C4)alkoxy;
R2 is _NR
aC(0)0(Ci-C4)alkyl; -NRaC(0)NRa(Ci-C4)alkyl; -NRaC(0)0(C2-
C4)alkenyl;
-NRaC(0)NRa(C2-C4)a1keny1; -NRaC(0)0-(C3-C6)cyc10a1ky1; -NRaC(0)NRa-(C3-
C7)cycloalkyl; -NRaC(0)0-pheny1; -NRaC(0)NRa-pheny1; -NRaC(0)0-monocyc1ic 3-7
membered heterocyclic ring; -NRaC(0)NRa-monocyc1ic 3-7 membered heterocyclic
ring; -
NRaC(0)0-monocyc1ic 5-6 membered heteroaromatic ring; -NRaC(0)NRa-monocyc1ic 5-
6
membered heteroaromatic ring;
wherein the (Ci¨C4)alkyl and the (C2-C4)alkenyl in the group represented by R2
are
each optionally and independently substituted with one or more groups selected
from the
group consisting of halogen, N3, ¨0Ra, -NRaRa, -(C3¨C6)cycloalkyl, phenyl, a
monocyclic 3-
7 membered heterocyclic ring, and a monocyclic 5-6 membered heteroaromatic
ring;
wherein the (C3¨C7)cycloalkyl in the group represented by R2 is optionally
substituted
with one or more groups selected from the group consisting of halogen, -CH3,
=0, -0Ra and
-NRaRa;
wherein the phenyl in the group represented by R2 is optionally substituted
with one
or more groups selected from the group consisting of halogen, -CH3,
halomethyl,
halomethoxy,
-CN, -0Ra, and -N3;
wherein the heterocyclic ring in the group represented by R2 is optionally
substituted
with one or more groups selected from the group consisting of =0, halogen,
¨0Ra, -CH3,
halomethyl, and halomethoxy;
wherein the heteroaromatic ring in the group represented by R2 is optionally
substituted with one or more groups selected from the group consisting of
halogen, -CN, -
CH3, halomethyl, halomethoxy, -0Ra and -NRaRa; and
each Ra is independently ¨H or -CH3, and
a PARP inhibitor or a pharmaceutically acceptable salt thereof
110

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
8. The kit of claim 7, wherein the PARP inhibitor is olaparib.
9. The kit of claim 7, wherein the compound of Formula (I) has the
following structure:
0
0
0
OAN S=0
HNI
10. A method of treating cancer, an autoimmune disease, an immune
deficiency, or a
neurodegenerative disease, comprising administering to a subject in need
thereof a
therapeutically effective amount of the combination therapy of any one of
claims 1-3, the
pharmaceutical composition of any one of claims 4-6, or the kit of any one of
claims 7-9.
11. A method of treating cancer, an autoimmune disease, an immune
deficiency, or a
neurodegenerative disease, comprising administering to a subject in need
thereof a
therapeutically effective amount of a compound represented by Formula (I):
0 R3
R2 S , \ X5Lxµ
HN -0
Formula (I);
or a pharmaceutically acceptable salt thereof, wherein:
the thiazole ring is optionally substituted with ¨F or ¨C1;
Cy is -(C3¨C7)cycloalkyl, bridged (C6¨C12) cycloalkyl, or a 4-12 membered
heterocyclic ring, each of which is optionally substituted with one or more
groups selected
from the group consisting of halogen, -OH, (C1-C4)alkyl, and (C1-C4)alkoxy;
when X5 is connected with a nitrogen ring atom of Cy, X5 is absent;
when X5 is connected with a carbon ring atom of Cy, X5 is NRa or 0;
X6 NRa or 0;
is (C1¨Cs)alkyl;
111

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
R3 is (C1¨05)alkyl, -CH2-phenyl, -(C3¨C7)cycloalkyl, -CH2-(C3¨C7)cycloalkyl,
-CH2-monocyclic 3-7 membered heterocyclic ring, or monocyclic 3-7 membered
heterocyclic
ring, wherein the (Ci¨Cs)alkyl, -(C3¨C7)cycloalkyl, phenyl or monocyclic 3-7
membered
heterocyclic ring represented by R3 or in the group represented by R3 is
optionally substituted
with one or more groups selected from the group consisting of halogen, -OH,
(Ci-C4)alkyl,
halomethyl, halomethoxy, -CN, and (Ci-C4)alkoxy;
R2 is _NR
aC(0)0(Ci-C4)alkyl; -NRaC(0)NRa(Ci-C4)alkyl; -NRaC(0)0(C2-
C4)alkenyl;
-NRaC(0)NRa(C2-C4)a1keny1; -NRaC(0)0-(C3-C6)cyc10a1ky1; -NRaC(0)NRa-(C3-
C7)cycloalkyl; -NRaC(0)0-pheny1; -NRaC(0)NRa-pheny1; -NRaC(0)0-monocyc1ic 3-7
membered heterocyclic ring; -NRaC(0)NRa-monocyc1ic 3-7 membered heterocyclic
ring; -
NRaC(0)0-monocyc1ic 5-6 membered heteroaromatic ring; -NRaC(0)NRa-monocyc1ic 5-
6
membered heteroaromatic ring;
wherein the (Ci¨C4)alkyl and the (C2-C4)alkenyl in the group represented by R2
are
each optionally and independently substituted with one or more groups selected
from the
group consisting of halogen, N3, ¨0Ra, -NRaRa, -(C3¨C6)cycloalkyl, phenyl, a
monocyclic 3-
7 membered heterocyclic ring, and a monocyclic 5-6 membered heteroaromatic
ring;
wherein the (C3¨C7)cycloalkyl in the group represented by R2 is optionally
substituted
with one or more groups selected from the group consisting of halogen, -CH3,
=0, -0Ra and
-NRaRa;
wherein the phenyl in the group represented by R2 is optionally substituted
with one
or more groups selected from the group consisting of halogen, -CH3,
halomethyl,
halomethoxy,
-CN, -0Ra, and -N3;
wherein the heterocyclic ring in the group represented by R2 is optionally
substituted
with one or more groups selected from the group consisting of =0, halogen,
¨0Ra, -CH3,
halomethyl, and halomethoxy;
wherein the heteroaromatic ring in the group represented by R2 is optionally
substituted with one or more groups selected from the group consisting of
halogen, -CN, -
CH3, halomethyl, halomethoxy, -0Ra and -NRaRa; and
each Ra is independently ¨H or -CH3, and
a therapeutically effective amount of a PARP inhibitor or a pharmaceutically
acceptable salt thereof.
112

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
12. The method of claim 11, wherein the PARP inhibitor is olaparib.
13. The method of claim 11, wherein the compound of Formula (I) has the
following
structure:
0 ___________________________________________________
0
OAN
0
S=0
HNI
14. The method of any one of claims 10-13, wherein the cancer is a
hematological cancer
or a solid tumor.
15. The method of any one of claims 10-13, wherein the cancer is a
hematological cancer
selected from lymphoma, leukemia, and a plasma cell neoplasm.
16. The method of claim 15, wherein the lymphoma is a B-cell lymphoma.
17. The method of claim 16, wherein the B-cell lymphoma is diffuse large B-
cell
lymphoma.
18. The method of claim 15, wherein the lymphoma is mantle cell lymphoma.
19. The method of claim 15, wherein the lymphoma is Burkitt's Lymphoma.
20. The method of any one of claims 10-13, wherein the cancer is a solid
tumor.
21. The method of claim 20, wherein the solid tumor is selected from breast
cancer,
ovarian cancer, pancreatic cancer, prostate cancer, fallopian tube cancer,
peritoneal cancer,
and lung cancer.
113

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
22. Use of a compound represented by Formula (I):
\ 0 R3
,o S 5L xµ
R2
s
H N \ 0
Formula (I);
or a pharmaceutically acceptable salt thereof, wherein:
the thiazole ring is optionally substituted with ¨F or ¨C1;
Cy is -(C3¨C7)cycloalkyl, bridged (C6¨C12) cycloalkyl, or a 4-12 membered
heterocyclic ring, each of which is optionally substituted with one or more
groups selected
from the group consisting of halogen, -OH, (C1-C4)alkyl, and (C1-C4)alkoxy;
when X5 is connected with a nitrogen ring atom of Cy, X5 is absent;
when X5 is connected with a carbon ring atom of Cy, X5 is NRa or 0;
X6 is NRa or 0;
le is (Ci¨Cs)alkyl;
R3 is (Ci¨Cs)alkyl, -CH2-phenyl, -(C3¨C7)cycloalkyl, -CH2-(C3¨C7)cycloalkyl,
-CH2-monocyclic 3-7 membered heterocyclic ring, or monocyclic 3-7 membered
heterocyclic
ring, wherein the (Ci¨Cs)alkyl, -(C3¨C7)cycloalkyl, phenyl or monocyclic 3-7
membered
heterocyclic ring represented by R3 or in the group represented by R3is
optionally substituted
with one or more groups selected from the group consisting of halogen, -OH,
(C1-C4)alkyl,
halomethyl, halomethoxy, -CN, and (C1-C4)alkoxy;
R2 is _NR
aC(0)0(C1-C4)alkyl; -NRaC(0)NRa(C1-C4)alkyl; -NRaC(0)0(C2-
C4)alkenyl;
-NRaC(0)NRa(C2-C4)a1keny1; -NRaC(0)0-(C3-C6)cyc1oa1ky1; -NRaC(0)NRa-(C3-
C7)cycloalkyl; -NRaC(0)0-pheny1; -NRaC(0)NRa-pheny1; -NRaC(0)0-monocyc1ic 3-7
membered heterocyclic ring; -NRaC(0)NRa-monocyc1ic 3-7 membered heterocyclic
ring; -
NRaC(0)0-monocyc1ic 5-6 membered heteroaromatic ring; -NRaC(0)NRa-monocyc1ic 5-
6
membered heteroaromatic ring;
wherein the (C1¨C4)alkyl and the (C2-C4)alkenyl in the group represented by R2
are
each optionally and independently substituted with one or more groups selected
from the
group consisting of halogen, N3, ¨0Ra, -NRaRa, -(C3¨C6)cycloalkyl, phenyl, a
monocyclic 3-
7 membered heterocyclic ring, and a monocyclic 5-6 membered heteroaromatic
ring;
114

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
wherein the (C3¨C7)cycloalkyl in the group represented by R2 is optionally
substituted
with one or more groups selected from the group consisting of halogen, -CH3,
=0, -0Ra and
_NRaRa;
wherein the phenyl in the group represented by R2 is optionally substituted
with one
or more groups selected from the group consisting of halogen, -CH3,
halomethyl,
halomethoxy,
-CN, -0Ra, and -N3;
wherein the heterocyclic ring in the group represented by R2 is optionally
substituted
with one or more groups selected from the group consisting of =0, halogen,
¨0Ra, -CH3,
halomethyl, and halomethoxy;
wherein the heteroaromatic ring in the group represented by R2 is optionally
substituted with one or more groups selected from the group consisting of
halogen, -CN, -
CH3, halomethyl, halomethoxy, -0Ra and -NRaRa; and
each Ra is independently ¨H or -CH3,
in combination with a PARP inhibitor or a pharmaceutically acceptable salt
thereof, in the
manufacture of a medicament for the treatment of a cancer, an autoimmune
disease, an
immune deficiency, or a neurodegenerative disease.
23. A compound represented by Formula (I):
\ 0 R3
R2 410 s
H N 0
Formula (I);
or a pharmaceutically acceptable salt thereof, wherein:
the thiazole ring is optionally substituted with ¨F or ¨C1;
Cy is -(C3¨C7)cycloalkyl, bridged (C6¨C12) cycloalkyl, or a 4-12 membered
heterocyclic ring, each of which is optionally substituted with one or more
groups selected
from the group consisting of halogen, -OH, (C1-C4)alkyl, and (C1-C4)alkoxy;
when X5 is connected with a nitrogen ring atom of Cy, X5 is absent;
when X5 is connected with a carbon ring atom of Cy, X5 is NRa or 0;
X6 is NRa or 0;
le is (C1¨Cs)alkyl;
115

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
R3 is (C1¨05)alkyl, -CH2-phenyl, -(C3¨C7)cycloalkyl, -CH2-(C3¨C7)cycloalkyl,
-CH2-monocyclic 3-7 membered heterocyclic ring, or monocyclic 3-7 membered
heterocyclic
ring, wherein the (Ci¨Cs)alkyl, -(C3¨C7)cycloalkyl, phenyl or monocyclic 3-7
membered
heterocyclic ring represented by R3 or in the group represented by R3 is
optionally substituted
with one or more groups selected from the group consisting of halogen, -OH,
(Ci-C4)alkyl,
halomethyl, halomethoxy, -CN, and (Ci-C4)alkoxy;
R2 is _NR
aC(0)0(Ci-C4)alkyl; -NRaC(0)NRa(Ci-C4)alkyl; -NRaC(0)0(C2-
C4)alkenyl;
-NRaC(0)NRa(C2-C4)a1keny1; -NRaC(0)0-(C3-C6)cyc10a1ky1; -NRaC(0)NRa-(C3-
C7)cycloalkyl; -NRaC(0)0-pheny1; -NRaC(0)NRa-pheny1; -NRaC(0)0-monocyc1ic 3-7
membered heterocyclic ring; -NRaC(0)NRa-monocyc1ic 3-7 membered heterocyclic
ring; -
NRaC(0)0-monocyc1ic 5-6 membered heteroaromatic ring; -NRaC(0)NRa-monocyc1ic 5-
6
membered heteroaromatic ring;
wherein the (Ci¨C4)alkyl and the (C2-C4)alkenyl in the group represented by R2
are
each optionally and independently substituted with one or more groups selected
from the
group consisting of halogen, N3, ¨0Ra, -NRaRa, -(C3¨C6)cycloalkyl, phenyl, a
monocyclic 3-
7 membered heterocyclic ring, and a monocyclic 5-6 membered heteroaromatic
ring;
wherein the (C3¨C7)cycloalkyl in the group represented by R2 is optionally
substituted
with one or more groups selected from the group consisting of halogen, -CH3,
=0, -0Ra and
-NRaRa;
wherein the phenyl in the group represented by R2 is optionally substituted
with one
or more groups selected from the group consisting of halogen, -CH3,
halomethyl,
halomethoxy,
-CN, -0Ra, and -N3;
wherein the heterocyclic ring in the group represented by R2 is optionally
substituted
with one or more groups selected from the group consisting of =0, halogen,
¨0Ra, -CH3,
halomethyl, and halomethoxy;
wherein the heteroaromatic ring in the group represented by R2 is optionally
substituted with one or more groups selected from the group consisting of
halogen, -CN, -
CH3, halomethyl, halomethoxy, -0Ra and -NRaRa; and
each Ra is independently ¨H or -CH3,
116

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
for use in combination with a PARP inhibitor or a pharmaceutically acceptable
salt thereof, in
treating a cancer, an autoimmune disease, an immune deficiency, or a
neurodegenerative
disease.
117

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
COMBINATIONS OF RAD51 AND PARP INHIBITORS
RELATED APPLICATIONS
This application claims priority to and the benefit of U.S. Provisional
Application No.
62/823,556, filed on March 25, 2019, the entire contents of which are
incorporated herein by
reference.
BACKGROUND
RAD51 is a member of the RAD51 family which promotes the repair of DNA double
strand breaks (DSB). RAD51 protein is highly conserved in most eukaryotes,
from yeast to
humans. The human RAD51 is a 339-amino acid protein that plays a major role in
DNA
replication and repair by homologous recombination (HR). RAD51 catalyzes
strand transfer
between a broken sequence and undamaged homologous template to allow re-
synthesis of the
damaged region.
Studies have demonstrated sensitization to certain DNA damaging therapies
associated with defects in proteins that promote HR DNA repair. This
sensitization is
particularly dramatic for DNA cross-linking chemotherapeutic drugs and
ionizing radiation.
It has been shown that HR can be partially inhibited in order to sensitize
cells to DNA
damaging therapies. For example, inhibition of XRCC3 (a RAD51 paralog protein)
using a
synthetic peptide sensitized Chinese Hamster Ovary (CHO) cells to cisplatin
and inhibited the
formation of sub-nuclear RAD51 foci in response to DNA damage. Researchers
have
inhibited the expression of the RAD51 protein itself or blocked its function
by over-
expressing a dominant negative BRC peptide fragment derived from BRCA2. In
view of the
connection between increased sensitivity to DNA damaging therapies and defects
in HR
DNA repair-related proteins, there is a need for compounds that inhibit RAD51.
Poly(ADIP-ribose) polyinerase (PARP) constitutes a super family of eighteen
proteins
containing PARP catalytic domains. These proteins include PARP-1, PARP-2, PARP-
3,
tatikyra.se-1, tankyra,se-2, vaultPARP and 71IPARP. PARP-1, the founding
member, consists
of three main domains: a N-terminal DNA-binding domain containing two zinc
fingers, an
automodifi cad on domain, and a C-terminal catalytic domain. PARP are nuclear
and
cytoplasmic enzymes that cleave NAD'to nicotinamide and ADP-ribose to form
long and
branched A.DP-ribose polymers on target proteins, includin.g topoisornerases,
liisiones, and.
PARP
1

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
Poly(ADP-ribosyflati on has been implicated in several biolo.fical processes,
including
DNA repair, gene transcription, cell cycle progression, cell death, chromatin
functions, and.
genomic stability. Inhibition of PARP has been shown to play a role in several
disease states.
PARP inhibitors have been used in prophylactic therapy for elimination of
BRCA2-deficient
cells, treatment of inflammation diseases, acute and chronic myocardial
diseases, vascular
diseases, septic shock, ischernic injury, and neurotoxicity, and treatment or
prevention of
autoimmune diseases such as Type I diabetes and diabetic complications. PARP
has also
been demonstrated to play a role in the pathogenesis of hemorrhagic shock.
Thus, there is a need for therapies that utilize both RA.D5l inhibitors and
PARP
inhibitors. The present application addresses the need.
SUMMARY
The present application discloses that inhibitors of RAD51 (see Compounds 1-
75,
Table 1) act synergistically in combination with PARP inhibitors. The present
application
provides a compound represented by Structural Formula I:
\ 0 R3
R2 /OS 0 x5L- X6/
,S
HN
or a pharmaceutically acceptable salt thereof, for use in combination with
PARP inhibitors.
The definition of each variable is provided below.
The present application also provides a pharmaceutical composition comprising
a
compound as described herein or a pharmaceutically acceptable salt thereof and
a
pharmaceutically acceptable carrier or diluent.
The present application provides a method of treating a cancer, an autoimmune
disease, an immune deficiency, or a neurodegenerative disease. The method
comprises
administering to a subject in need thereof a therapeutically effective amount
of a compound
disclosed herein or a pharmaceutically acceptable salt thereof or a
pharmaceutical
composition disclosed herein.
The present application provides the use of a compound disclosed herein, or a
pharmaceutically acceptable salt thereof, or a pharmaceutical composition
disclosed herein in
the manufacture of a medicament for the treatment of a cancer, an autoimmune
disease, an
immune deficiency, or a neurodegenerative disease.
2

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
The present application provides a compound disclosed herein, or a
pharmaceutically
acceptable salt thereof, or a pharmaceutical composition disclosed herein for
use in treating a
cancer, an autoimmune disease, an immune deficiency, or a neurodegenerative
disease.
The present application provides a method of treating a cancer, an autoimmune
disease, an immune deficiency, or a neurodegenerative disease, wherein the
method
comprises administering to a subject in need thereof a therapeutically
effective amount of a
compound disclosed herein or a pharmaceutically acceptable salt thereof or a
pharmaceutical
composition disclosed herein in combination with a PARP inhibitor or a
pharmaceutically
acceptable salt thereof or a pharmaceutical composition comprising a PARP
inhibitor.
The present application provides use of a compound disclosed herein, or a
pharmaceutically acceptable salt thereof, or a pharmaceutical composition
disclosed herein in
combination with a PARP inhibitor, or a pharmaceutically acceptable salt
thereof, or a
pharmaceutical composition thereof in the manufacture of a medicament for the
treatment of
a cancer, an autoimmune disease, an immune deficiency, or a neurodegenerative
disease.
The present application provides a compound disclosed herein, or a
pharmaceutically
acceptable salt thereof, or a pharmaceutical composition disclosed herein in
combination with
a PARP inhibitor, or a pharmaceutically acceptable salt thereof, or a
pharmaceutical
composition thereof for use in treating a cancer, an autoimmune disease, an
immune
deficiency, or a neurodegenerative disease.
The present application provides a method of treating a cancer, an autoimmune
disease, an immune deficiency, or a neurodegenerative disease. The method
comprises
administering to a subject in need thereof a composition comprising a compound
disclosed
herein and a PARP inhibitor.
The present application provides a kit comprising a compound disclosed herein,
or a
pharmaceutically acceptable salt thereof, or a pharmaceutical composition
disclosed herein,
and a PARP inhibitor, or a pharmaceutically acceptable salt thereof, or a
pharmaceutical
composition thereof.
In some embodiments, a compound disclosed herein and the PARP inhibitor are
administered in temporal proximity for treating cancer. In some embodiments,
the application
provides a synergistic composition of the compound disclosed herein and the
PARP inhibitor,
wherein the compound disclosed herein and the PARP inhibitor come into contact
with each
other in the human body (e.g., only in the human body).
3

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
In some embodiments, the application provides a method of preparing a
composition
by bringing the compound disclosed herein and the PARP inhibitor into contact
with each
other at a locus.
BRIEF DESCRIPTION OF THE FIGURES
The patent or application. file contains at least one drawing executed in
color. Copies
of this patent or patent application publication with color drawing(s) will be
provided by the
Office upon request and payment of the necessary fee.
Bar colors represent particular concentrations of Compound # (e.g., Compound
67A).
Dark purple represents 19.53.125 TIM, dark blue represents 39.0625 nM, brown
represents
78.125 nM, black represents 156.25 nM, orange represents 312.5 nM, light
purple represents
625 lira& green represents 1250 niVI, red represents 250011M and light blue
represents 5000
nM.
FIG. 1A shows rucaparib - Compound 67A combination studies. Cells treated were

ARPE19/HPV16 cells. Bar chart is Bliss Independence data plotted, and heatmap
is
according to Loewe additivity model.
FIG. 1B shows rucaparib - Compound 67A combination studies. Cells treated were

Daudi cells. Bar chart is Bliss Independence data plotted, and heatmap is
according to Loewe
additivity model.
FIG. 1C shows rucaparib - Compound 67A combination studies. Cells treated were

ARPE19/HPV16 cells. Bar chart is Bliss Independence data plotted, and heatmap
is
according to Loewe additivity model.
FIG. 2A shows talazoparib - Compound 67A combination studies. Cells treated
were
ARPE19/HPV16 cells. Bar chart is Bliss Independence data plotted, and heatmap
is
according to Loewe additivity model.
FIG. 2B shows talazoparib - C:ompound 67A combination studies. Cells treated
were
Daudi cells. Bar chart is Bliss Independence data plotted, and heatmap is
according to Loewe
additivity model.
FIG. 2C shows talazoparib - Compound 67A combination studies. Cells treated
were
ARPE19/HPV16 cells. Bar chart is Bliss Independence data plotted, and heatmap
is
according to Loewe additivity model.
4

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
FIG. 3A shows nirapairb - Compound 67A combination studies. Cells treated were

ARPE19/HPV16 cells. Bar chart is Bliss Independence data plotted, and heatmap
is
according to Loewe additivity model.
FIG. 3B shows nirapairb - Compound 67A combination studies. Cells treated were

Daudi cells. Bar chart is Bliss Independence data plotted, and heatmap is
according to Loewe
additivity model.
FIG. 3C shows nirapairb - Compound 67A combination studies. Cells treated were

ARPE19/HPV16 cells. Bar chart is Bliss Independence data plotted, and heatmap
is
according to Loewe additivity model.
FIG. 4A shows olaparib - Compound 67A combination studies. Cells treated were
ARPE19/HPV16 cells. Bar chart is Bliss Independence data plotted, and heatmap
is
according to Loewe additivity model.
FIG. 4B shows olaparib - Compound 67A combination studies. Cells treated were
Daudi cells. Bar chart is Bliss Independence data plotted, and heatmap is
according to Loewe
additivity model.
FIG. 4C shows olaparib - Compound 67A combination studies. Cells treated were
KYSE-70 cells. Bar chart is Bliss Independence data plotted, and heatmap is
according to
Loewe additivity model.
FIG. 4D shows olaparib - Compound 67A combination studies. Cells treated were
BT-20 cells. Bar chart is Bliss Independence data plotted, and heatmap is
according to Loewe
additivity model.
FIG. 4E shows olaparib - Compound 67A combination studies. Cells treated were
HCC1937 cells. Bar chart is Bliss Independence data plotted, and heatmap is
according to
Loewe additivity model.
FIG. 5A shows veliparib - Compound 67A combination studies. Cells treated were

ARPE19/HPV16 cells. Bar chart is Bliss Independence data plotted, and heatmap
is
according to Loewe additivity model.
FIG. 5B shows veliparib - Compound 67A combination studies. Cells treated were

Daudi cells. Bar chart is Bliss Independence data plotted, and heatmap is
according to Loewe
additivity model.
FIG. 5C shows veliparib - Compound 67A combination studies. Cells treated were

KYSE-70 cells. Bar chart is Bliss Independence data plotted, and heatmap is
according to
Loewe additivity model.

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
FIG. 5D shows veliparib - Compound 67A combination studies. Cells treated were

HCC1143 cells. Bar chart is Bliss Independence data plotted, and heatmap is
according to
Loewe additivity model.
FIG. 5E shows olaparib - Compound 67A combination studies. Cells treated were
HCC1937 cells. Bar chart is Bliss Independence data plotted, and heatmap is
according to
Loewe additivity model.
FIG. 6 shows a barchart key denoting concentration of Compound 67A and the
experimental setup.
FIG. 7A depicts tumor volume change over day with olaparib, Compound 67A, and
a combination thereof in a HBCx11 model.
FIG. 7B depicts TGI% with olaparib, Compound 67A, and a combination thereof in

a HBCx11 model.
FIG. 8A depicts tumor volume change over day with olaparib, Compound 67A, and
a combination thereof in a HBCx1 model.
FIG. 8B depicts TGI% with olaparib, Compound 67A, and a combination thereof in

a HBCx1 model.
FIG. 9A depicts tumor volume change over day with olaparib, Compound 67A, and
a combination thereof in a TM01079 model.
FIG. 9B depicts TGI% with olaparib, Compound 67A, and a combination thereof in

a TM01079 model.
FIG. 10A depicts tumor volume change over day with olaparib, Compound 67A, and

a combination thereof in a TM00090 cells.
FIG. 10B depicts TGI% with olaparib, Compound 67A, and a combination thereof
in
a TM00090 model.
FIG. 11A depicts tumor volume change over day with olaparib, Compound 67A, and

a combination thereof in a HBCx27 model.
FIG. 11B depicts TGI% with olaparib, Compound 67A, and a combination thereof
in
a HBCx27 model.
FIG. 12A depicts tumor volume change over day with olaparib, Compound 67A, and

a combination thereof in a HBCx12B model.
FIG. 12B depicts TGI% with olaparib, Compound 67A, and a combination thereof
in
a HBCx12B model.
6

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
FIG. 13A depicts tumor volume change over day with olaparib, Compound 67A, and

a combination thereof in a TM00091 model.
FIG. 13B depicts TGI% with olaparib, Compound 67A, and a combination thereof
in
a TM00091 model.
DETAILED DESCRIPTION
In a first embodiment, the application provides a compound represented by
Structural
Formula I in combination with a PARP inhibitor:
, N
/ \ 0 R3
R2 ,o S 0 LA
x5
HN µ0
I;
or a pharmaceutically acceptable salt thereof, wherein:
the thiazole ring is optionally substituted with ¨F or ¨Cl;
Cy is -(C3¨C7)cycloalkyl, bridged (C6¨C12) cycloalkyl, or a 4-12 membered
heterocyclic ring, each of which is optionally substituted with one or more
groups selected
from the group consisting of halogen, -OH, (C1-C4)alkyl, and (C1-C4)alkoxy;
when X5 is connected with a nitrogen ring atom of Cy, X5 is absent;
when X5 is connected with a carbon ring atom of Cy, X5 is NRa or 0;
X6 is NRa or 0;
RI- is (Ci¨05)alkyl;
R3 is (Ci¨05)alkyl, -CH2-phenyl, -(C3¨C7)cycloalkyl, -CH2-(C3¨C7)cycloalkyl,
-CH2-monocyclic 3-7 membered heterocyclic ring, or monocyclic 3-7 membered
heterocyclic
ring, wherein the (Ci¨05)alkyl, -(C3¨C7)cycloalkyl, phenyl or monocyclic 3-7
membered
heterocyclic ring represented by R3 or in the group represented by R3 is
optionally substituted
with one or more groups selected from the group consisting of halogen, -OH,
(C1-C4)alkyl,
halomethyl, halomethoxy, -CN, and (C1-C4)alkoxy;
R2 is
INK u(0)0(C1-C4)alkyl; -NRaC(0)NRa(C1-C4)alkyl; -NRaC(0)0(C2-
C4)alkenyl; -NRaC(0)NRa(C2-C4)alkenyl; -NRaC(0)0-(C3-C6)cycloalkyl; -
NRaC(0)NRa-(C3-
C7)cycloalkyl; -NRaC(0)0-phenyl; -NRaC(0)NRa-phenyl; -NRaC(0)0-monocyclic 3-7
membered heterocyclic ring; -NRaC(0)NRa-monocyclic 3-7 membered heterocyclic
ring; -
7

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
ac(o)0-monocyclic 5-6 membered heteroaromatic ring; -NRaC(0)NRa-monocyclic 5-6

membered heteroaromatic ring;
wherein the (C1-C4)alkyl and the (C2-C4)alkenyl in the group represented by R2
are
each optionally and independently substituted with one or more groups selected
from the
group consisting of halogen, N3, -OR, -NRalta, -(C3-C6)cycloalkyl, phenyl, a
monocyclic 3-
7 membered heterocyclic ring, and a monocyclic 5-6 membered heteroaromatic
ring;
wherein the (C3-C7)cycloalkyl in the group represented by R2 is optionally
substituted
with one or more groups selected from the group consisting of halogen, -CH3,
=0, -0Ra and
_NRaRa;
wherein the phenyl in the group represented by R2 is optionally substituted
with one
or more groups selected from the group consisting of halogen, -CH3,
halomethyl,
halomethoxy,
-CN, -0Ra, and -N3;
wherein the heterocyclic ring in the group represented by R2 is optionally
substituted
with one or more groups selected from the group consisting of =0, halogen, -
0Ra, -CH3,
halomethyl, and halomethoxy;
wherein the heteroaromatic ring in the group represented by R2 is optionally
substituted with one or more groups selected from the group consisting of
halogen, -CN, -
CH3, halomethyl, halomethoxy, -0Ra and -NRaRa; and
each Ra is independently -H or -CH3.
In a second embodiment, the application provides a compound represented by
Structural Formula II in combination with a PARP inhibitor:
/ N 0 R3
R2 goo ,Os x5Lx6
HN \13
R1 II;
or a pharmaceutically acceptable salt thereof, wherein
the thiazole ring is optionally substituted with -F or -Cl;
Cy is cyclohexyl or a 6-membered monocyclic heterocyclic ring;
X5 and X6 are each independently NR a or 0;
R1 is (Ci-05)alkyl;
R3 is (Ci-05)alkyl or monocyclic 3-7-membered heterocyclic ring;
8

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
R2 is
1NIC u(0)0(C1-C4)alkyl; -NRaC(0)NRa(C1-C4)alkyl; -NRaC(0)0(C2-
C4)alkenyl; -NRaC(0)NRa(C2-C4)alkenyl; -NRaC(0)-0(C3-C6)cycloalkyl; -
NRaC(0)NRa-(C3-
C6)cycloalkyl; -NRaC(0)0-phenyl; -NRaC(0)NRa-phenyl; -NRaC(0)0-monocyclic 3-7
membered heterocyclic ring; -NRaC(0)NRa-monocyclic 3-7 membered heterocyclic
ring; -
NRaC(0)0-monocyclic 5-6 membered heteroaromatic ring; -NRaC(0)NRa-monocyclic 5-
6
membered heteroaromatic ring;
wherein the (C1¨C4)alkyl and the (C2-C4)alkenyl in the group represented by R2
are
each optionally and independently substituted with one or more halogen, N3,
-NRaRa, -(C3¨C6)cycloalkyl, phenyl, monocyclic 3-7-membered heterocyclic ring,
or
monocyclic 5-6-membered heteroaromatic ring;
wherein the -(C3¨C6)cycloalkyl in the group represented by R2 is optionally
substituted with one or more halogen, -CH3, -0Ra or -NRaRa;
wherein the phenyl in the group represented by R2 is optionally substituted
with one
or more halogen, -CH3, halomethyl, halomethoxy, -0Ra, or -N3;
wherein the heterocyclic ring in the group represented by R2 is optionally
substituted
with one or more =0, halogen, -CH3, halomethyl, or halomethoxy;
wherein the heteroaromatic ring in the group represented by R2 is optionally
substituted with one or more halogen, -CH3, halomethyl, halomethoxy, -0Ra or -
NRaRa; and
each Ra is independently ¨H or -CH3.
In a third embodiment, the application provides a compound according to
Structural
Formula I, or a pharmaceutically acceptable salt thereof, in combination with
a PARP
inhibitor, wherein Cy is cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl,
cycloheptyl;
azetidinyl, azepanyl, diazaspiro[4.4]nonyl, diazaspiro[3.5]nonyl, diazepanyl,
dihydroimidazole, dihydrofuranyl, dihydropyranyl, dihydropyridinyl,
dihydropyrimidinyl,
dihydrothienyl, dihydrothiophenyl, dihydrothiopyranyl, hexahydropyridazinyl,
hexahydropyrimidinyl, hydantoinyl, indolinyl, isoindolinyl, morpholinyl,
oxiranyl, oxetanyl,
piperidinyl, piperazinyl, pyrrolidinonyl, pyrrolidinyl, tetrahydrofuranyl,
tetrahydroimidazole,
tetrahydroindolyl, tetrahydropyranyl, tetrahydrothienyl, tetrahydropyridinyl,
tetrahydropyrimidinyl, tetrahydrothiophenyl, tetrahydrothiopyranyl,
thiomorpholinyl,
tropanyl, valerolactamyl; bicyclo[2.1.1]hexyl, bicyclo[2.2.1]heptyl,
bicyclo[2.2.2]octyl,
bicyclo[3.2.1]octyl, bicyclo[4.3.1]decyl, bicyclo[3.3.1]nonyl, bornyl,
bornenyl, norbornyl,
norbornenyl, 6,6-dimethylbicyclo [3.1.1]heptyl, tricyclobutyl, adamantly;
azanorbornyl,
quinuclidinyl, isoquinuclidinyl, tropanyl, azabicyclo[2.2.1]heptanyl, 2-
9

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
azabicyclo[3.2.1]octanyl, azabicyclo[3.2.1]octanyl, azabicyclo[3.2.2]nonanyl,
azabicyclo[3.3.0]nonanyl, azabicyclo [3.3.1]nonanyl,
diazabicyclo[2.2.1]heptanyl,
diazabicyclo[3.2.1]octanyl, octahydropyrrolo[3,4-b]pyrrolyl,
octahydropyrrolo[3,4-
c]pyrroly1; and the remaining variables are as defined in the first
embodiment.
In a fourth embodiment, the application provides a compound according to
Structural
Formula I or II, or a pharmaceutically acceptable salt thereof, in combination
with a PARP
inhibitor, wherein Cy is cyclohexyl, morpholinyl, thiomorpholinyl,
piperidinyl, piperazinyl,
hexahydropyridazinyl, hexahydropyrimidinyl, valerolactamyl, dihydropyranyl,
dihydropyridinyl, dihydropyrimidinyl, dihydrothiopyranyl, tetrahydropyranyl,
tetrahydropyridinyl, tetrahydropyrimidinyl, or tetrahydrothiopyranyl; and the
remaining
variables are as defined in the first, second, and/or third embodiments.
In a fifth embodiment, the application provides a compound represented by
Structural
Formula III in combination with a PARP inhibitor:
, N
HN ,0 X5 's
X7'"
0 HN
141
or a pharmaceutically acceptable salt thereof, wherein:
X7 is NH or 0;
R4 is (C1¨C4)alkyl, (C3-C6)cycloalkyl, or a monocyclic 3-7 membered
heterocyclic
ring;
wherein the (C1¨C4)alkyl represented by R4 is optionally substituted with one
or more
R,
groups selected from the group consisting of halogen, N3, ¨0Ra, -NRaa
-(C3¨C6)cycloalkyl, phenyl, a monocyclic 3-7 membered heterocyclic ring, and a
monocyclic
5-6 membered heteroaromatic ring,
wherein the (C3-C6)cycloalkyl or the monocyclic 3-7 membered heterocyclic ring

represented by R4, the (C3-C6)cycloalkyl or the monocyclic 3-7 membered
heterocyclic ring
in the group represented by R4 is optionally substituted with one or more
groups selected
from the group consisting of halogen, -0Ra, =0, and -CH3,
wherein the phenyl in the group represented by R4 is optionally substituted
with one
or more groups selected from the group consisting of halogen, -CH3,
halomethyl,
halomethoxy, -0Ra, and -N3;

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
wherein the heteroaromatic ring in the group represented by R4 is optionally
substituted with one or more groups selected from the group consisting of
halogen and -CH3;
and the remaining variables are as defined in the first, second, third, and/or
fourth
embodiments.
In a sixth embodiment, the application provides a compound according to
Structural
Formula III, or a pharmaceutically acceptable salt thereof, in combination
with a PARP
inhibitor, wherein X' is NH or 0; R3 is (Ci¨05)alkyl; and le is (C1¨C4)alkyl
wherein the
(C1¨C4)alkyl represented by R4 is optionally substituted with one or more
halogen, ¨0Ra, -
NRa =-= a
, -(C3¨C6)cycloalkyl, phenyl (optionally substituted by one or more halogen, -
CH3,
halomethyl, halomethoxy, ORa or N3), monocyclic 3-7-membered heterocyclic ring

(optionally substituted by =0, halogen or ¨CH3), or monocyclic 5-6-membered
heteroaromatic ring (optionally substituted by halogen or ¨CH3); and the
remaining variables
are as defined in the first, second, third, fourth and/or fifth embodiments.
In a seventh embodiment, the application provides a compound represented by
Structural Formula IV in combination with a PARP inhibitor:
/
HN S)a 0
,0 u R3
,Sµ X5 X6
HN NO
RI 1
or a pharmaceutically acceptable salt thereof; and the variables are as
defined in the first,
second, third, fourth, fifth and/or sixth embodiments.
In an eighth embodiment, the application provides a compound represented by
Structural Formula V in combination with a PARP inhibitor:
/
HN S)C3lj x5 X6 0
X7 ,0 R3
,S ,
let HN \O
RI 1 V,
or a pharmaceutically acceptable salt thereof; and the variables are as
defined in the first,
second, third, fourth, fifth and/or sixth embodiments.
In a ninth embodiment, the application provides a compound represented by
Structural Formula VI in combination with a PARP inhibitor:
11

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
HN S) 0
R3
,S, X5 X6
144 HN NO
RI I VI;
or a pharmaceutically acceptable salt thereof; and the variables are as
defined in the first,
second, third, fourth, fifth and/or sixth embodiments.
In a tenth embodiment, the application provides a compound represented by
Structural
Formula VII in combination with a PARP inhibitor:
N
HN S 0
,0 /R3
X6
144 HN \O
RI I
or a pharmaceutically acceptable salt thereof; and the variables are as
defined in the first,
second, third, fourth, fifth and/or sixth embodiments.
In an eleventh embodiment, the application provides a compound represented by
Structural Formula VIII in combination with a PARP inhibitor:
HN S'CO 0
N.x511...x6/R3
144 HN NO
RI I
or a pharmaceutically acceptable salt thereof; and the variables are as
defined in the first,
second, third, fourth, fifth and/or sixth embodiments.
In a twelfth embodiment, the application provides a compound represented by
Structural Formula IX in combination with a PARP inhibitor:
N
/
HN 0
x5ILX6/R3
144 HN 0
R1
;
or a pharmaceutically acceptable salt thereof; and the variables are as
defined in the first,
second, third, fourth, fifth and/or sixth embodiments.
In a thirteenth embodiment, the application provides a compound according to
Structural Formula I, II, or III, or a pharmaceutically acceptable salt
thereof, in combination
with a PARP inhibitor, wherein Cy is azetidinyl or pyrrolidinyl, and the
nitrogen ring atom is
12

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
connected with the thiazole ring; and the remaining variables are as defined
in the first,
second, third, fourth, fifth and/or sixth embodiments.
In a fourteenth embodiment, the application provides a compound according to
Structural Formula I, II, or III, or a pharmaceutically acceptable salt
thereof, in combination
with a PARP inhibitor, wherein Cy is 1,7-diazaspiro[4.4]nonyl, 2,7-
diazaspiro[4.4]nonyl, 2,7-
diazaspiro[3.5]nonyl, 1,4-diazepanyl, 2,5-diazabicyclo[2.2.1]heptanyl, 3,8-
diazabicyclo[3.2.1]octanyl, octahydropyrrolo[3,4-b]pyrrolyl, or
octahydropyrrolo[3,4-
c]pyrrolyl, and the two nitrogen ring atoms are connected with the thiazole
ring and the -
X5C(0)X6R3 moiety, respectively; and the remaining variables are as defined in
the first,
second, third, fourth, fifth and/or sixth embodiments.
In a fifteenth embodiment, the application provides a compound according to
Structural Formula III, IV, V, VI, VII, VIII, or IX, or a pharmaceutically
acceptable salt
thereof, in combination with a PARP inhibitor, wherein R4 is -(C1¨C3)alkyl,
(C3-
C6)cycloalkyl, or a monocyclic 3-7 membered heterocyclic ring, wherein the -
(C1¨C3)alkyl is
optionally substituted with (i) phenyl optionally substituted by one or more
halogen or -CH3;
(ii) a monocyclic 5-6 membered heteroaromatic ring optionally substituted by
one or more
halogen or ¨CH3; or (iii) a monocyclic 3-7 membered heterocyclic ring
optionally substituted
by one or more groups selected from the group consisting of halogen and ¨CH3;
and the
remaining variables are as defined in the first, second, third, fourth, fifth,
seventh, eighth,
ninth, tenth, eleventh, twelfth, thirteenth, and/or fourteenth embodiments.
In a sixteenth embodiment, the application provides a compound according to
Structural Formula III, IV, V, VI, VII, VIII, or IX, or a pharmaceutically
acceptable salt
thereof, in combination with a PARP inhibitor, wherein R4 is -(C1¨C3)alkyl, -
CHRa-phenyl, -
CHRa-5-6 membered heteraromatic ring, or -CHRa-3-7 membered monocyclic
heterocyclic
ring, wherein the phenyl, 5-6 membered heteraromatic ring or 3-7 membered
monocyclic
heterocyclic ring in the group represented by R4 is optionally substituted one
or more groups
selected from the group consisting of halogen and ¨CH3; and the remaining
variables are as
defined in the first, second, third, fourth, fifth, seventh, eighth, ninth,
tenth, eleventh, twelfth,
thirteenth, and/or fourteenth embodiments.
In a seventeenth embodiment, the application provides a compound according to
Structural Formula III, IV, V, VI, VII, VIII, or IX, or a pharmaceutically
acceptable salt
thereof, in combination with a PARP inhibitor, wherein R4 is -(C1¨C3)alkyl,
optionally
substituted with (i) phenyl optionally substituted by one or more halogen, -
CH3, halomethyl,
13

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
halomethoxy, ORa, or N3; (ii) a monocyclic 5-6-membered heteroaromatic ring
optionally
substituted by one or more halogen or -CH3; or (iii) a monocyclic 3-7-membered

heterocyclic ring optionally substituted by one or more =0 or ¨CH3; and the
remaining
variables are as defined in the first, second, third, fourth, fifth, sixth,
seventh, eighth, ninth,
tenth, eleventh, twelfth, thirteenth, and/or fourteenth embodiments.
In an eighteenth embodiment, the application provides a compound according to
Structural Formula III, IV, V, VI, VII, VIII, or IX, or a pharmaceutically
acceptable salt
thereof, in combination with a PARP inhibitor, wherein R4 is -(C1¨C3)alkyl,
optionally
substituted with (i) phenyl optionally substituted by one or more halogen, -
CH3, halomethyl,
halomethoxy, ORa, or N3; (ii) a monocyclic 5-6-membered heteroaromatic ring
optionally
substituted by one or more halogen or -CH3; or (iii) a monocyclic 3-7-membered

heterocyclic ring optionally substituted by one or more =0 or ¨CH3; and the
remaining
variables are as defined in the first, second, third, fourth, fifth, sixth,
seventh, eighth, ninth,
tenth, eleventh, twelfth, thirteenth, fourteenth, and/or seventeenth
embodiments.
In a nineteenth embodiment, the application provides a compound according to
Structural Formula I, III, IV, V, VI, VII, VIII, or IX, or a pharmaceutically
acceptable salt
thereof, in combination with a PARP inhibitor, wherein R3 is (C1¨C4)alkyl, -
(C4¨C6)cycloalkyl, -CH2-phenyl, -CH2-monocyclic 4-6 membered heterocyclic
ring, or
monocyclic 4-6 membered heterocyclic ring, wherein the phenyl or monocyclic 4-
6
membered heterocyclic ring represented by R3 or in the group represented by R3
is optionally
substituted with one or more groups selected from the group consisting of
halogen,
-OR', and ¨CH3; and the remaining variables are as defined in the first,
third, fourth, fifth,
seventh, eighth, ninth, tenth, eleventh, twelfth, thirteenth, fourteenth,
fifteenth, sixteenth,
seventeenth, and/or eighteenth embodiments.
In a twentieth embodiment, the application provides a compound represented by
Structural Formula X in combination with a PARP inhibitor:
/
HN
NJ-L0/
144 HN \O
141 X;
or a pharmaceutically acceptable salt thereof; and the variables are as
defined in the first,
second, third, fourth, fifth, sixth, seventh, fifteenth, sixteenth,
seventeenth, eighteenth, and/or
nineteenth embodiments.
14

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
In a twenty first embodiment, the application provides a compound represented
by
Structural Formula XI in combination with a PARP inhibitor:
/
7sO
HN S 0
,0
144 HN NO ."N/R3
141
XI;
or a pharmaceutically acceptable salt thereof; and the variables are as
defined in the first,
second, third, fourth, fifth, sixth, seventh, fifteenth, sixteenth,
seventeenth, eighteenth, and/or
nineteenth embodiments.
In a twenty second embodiment, the application provides a compound represented
by
Structural Formula XII in combination with a PARP inhibitor:
i*Z)
HN S 0
141
or a pharmaceutically acceptable salt thereof; and the variables are as
defined in the first,
second, third, fourth, fifth, sixth, eighth, fifteenth, sixteenth,
seventeenth, eighteenth, and/or
nineteenth embodiments.
In a twenty third embodiment, the application provides a compound represented
by
Structural Formula XIII(a) or XIII(b) in combination with a PARP inhibitor:
HN S 0
)fin ,0
).L /R3
"'N 0
R4 - HN \ 0
141
XIII(a), or
HN /
/0
S; N )=0/ R3
144o HN, NO
141
XIII(b);
or a pharmaceutically acceptable salt thereof; and the variables are as
defined in the first,
second, third, fourth, fifth, sixth, eighth, fifteenth, sixteenth,
seventeenth, eighteenth, and/or
nineteenth embodiments.
In a twenty fourth embodiment, the application provides a compound represented
by
Structural Formula XIV in combination with a PARP inhibitor:

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
, N
HN N ,0 0
X7-co HN \
144
R1 XIV;
or a pharmaceutically acceptable salt thereof; and the variables are as
defined in the first,
second, third, fourth, fifth, sixth, tenth, fifteenth, sixteenth, seventeenth,
eighteenth, and/or
nineteenth embodiments.
In a twenty fifth embodiment, the application provides a compound according to

Structural Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII(a),
XIII(b), XIV, or
a pharmaceutically acceptable salt thereof, in combination with a PARP
inhibitor, wherein le
is isopropyl, tert-butyl, cyclobutyl, cyclopentyl, benzyl, oxetanyl,
tetrahydro-2H-pyranyl, or
NCE13 ; and the variables are as defined in the first, second, third,
fourth, fifth, sixth,
seventh, eighth, ninth, tenth, eleventh, twelfth, thirteenth, fourteenth,
fifteenth, sixteenth,
seventeenth, eighteenth, nineteenth, twentieth, twenty first, twenty second,
twenty third
and/or twenty fourth embodiments. In an alternative embodiment, le is
isopropyl or oxetanyl.
In another alternative embodiment, le is isopropyl.
In a twenty sixth embodiment, the application provides a compound according to

Structural Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII(a),
XIII(b), XIV, or
a pharmaceutically acceptable salt thereof, in combination with a PARP
inhibitor, wherein le
is tert-butyl; and the variables are as defined in the first, second, third,
fourth, fifth, sixth,
seventh, eighth, ninth, tenth, eleventh, twelfth, thirteenth, fourteenth,
fifteenth, sixteenth,
seventeenth, eighteenth, nineteenth, twentieth, twenty first, twenty second,
twenty third,
twenty fourth, and/or twenty fifth embodiments.
In a twenty seventh embodiment, the application provides a compound according
to
Structural Formula III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII(a),
XIII(b), XIV, or a
pharmaceutically acceptable salt thereof, in combination with a PARP
inhibitor, wherein R4
H 3C ?
is CH3 , N,r5 ELS , 4111
16

CA 03133005 2021-09-08
WO 2020/198298
PCT/US2020/024601
N
el N N
N y Iz. AJk
F
N
N NC)-- \PPPF HN
N )- N A ,or CH3 ; ana' the variables
are
as defined in the first, second, third, fourth, fifth, sixth, seventh, eighth,
ninth, tenth, eleventh,
twelfth, thirteenth, fourteenth, fifteenth, sixteenth, seventeenth,
eighteenth, nineteenth,
twentieth, twenty first, twenty second, twenty third, twenty fourth, twenty
fifth, and/or twenty
H3c,TA 0
sixth embodiments. In an alternative embodiment, le is cH3
F
el N
I HN
CNI>71Thrri4 , or bH3 .
In
F F 7
7
:
another alternative embodiment, le is el 1.1 lei I.
, , , ,
N Nn :
1 1 ' . c.....õ%,\ N
,
NCI--....\1 / NH / NC H 3 ,or CH3 . In
, ,
=
:
another alternative embodiment, le is 10 el Q Nc H3 T.miNi
rssr r rµ-''.16\6sss
, , ,
Cro\ N oss H 3c ,TA
NH N
csss , or C H
3 . Still in another alternative embodiment, le is
F =
.`N
y.),. I. /N ..sy N scss
0 , F I
, or I
. Still in another
H3c,y,µ 0
alternative embodiment, le is cH3 or .
The present application provides a compound represented by Structural Formula
I' in
combination with a PARP inhibitor.
In a first embodiment, the application provides a compound represented by
Structural
Formula I' in combination with a PARP inhibitor:
17

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
N
\ 0
Re 0 )=\ R3
X5 )K
(Rd) X4 HN ,SVD
HN NO
R1 I';
or a pharmaceutically acceptable salt thereof, wherein:
the thiazole ring is optionally substituted with -F or ¨Cl;
X4 is NRa or 0;
X5 and X6 are each independently NRb or 0;
R' is (Ci¨05)alkyl;
R3 is (Ci¨05)alkyl, -(C3¨C7)cycloalkyl, or ¨(CH2)qheterocycly1 (wherein the
heterocycyl is a monocyclic 3-7-membered heterocyclic ring optionally
substituted with one
or more occurences of methyl), or benzyl (wherein the benzyl ring is
optionally substituted
with one or more occurences of halogen, methoxy, halomethoxy, methyl,
halomethyl, or
cyano);
each of Ra, Rb, and RC is independently hydrogen or methyl;
Rd is independently halogen, methoxy, halomethoxy, methyl, halomethyl, or
cyano;
m is 0, 1, 2, or 3;
n is 0, 1, or 2; and
q is 0 or 1.
In a second embodiment, the application provides a compound represented by
Structural Formula I'-1 in combination with a PARP inhibitor:
N
\ 0
Re 0 ,R3
N 0
(Rd)m4 X4 HN 7s0
HN \O
R1 PA;
or a pharmaceutically acceptable salt thereof, and the variables are as
defined in the first
embodiment.
In a third embodiment, the application provides a compound represented by
Structural
Formula I'-2 in combination with a PARP inhibitor:
18

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
a 0
Rc 0 S )\ 7R3
J,L N 0
(Rd)m¨F- X4 HN H
HN \O
R1 I'-2;
or a pharmaceutically acceptable salt thereof, and the variables are as
defined in the first
embodiment.
In a forth embodiment, the application provides a compound represented by
Structural
Formula I'-3 in combination with a PARP inhibitor:
N
/ 0
Rc 0 S )\ 7R3
J,L IN 0
(Rd)m4 X4 HN ,S"\-=- H
HN \O
R1 I'-3;
or a pharmaceutically acceptable salt thereof, and the variables are as
defined in the first
embodiment.
In a fifth embodiment, the application provides a compound represented by
Structural
Formula I'-4 in combination with a PARP inhibitor:
0
Rc 0 S R3
N 0
(Rd)m4 X4 HN H
HN \O
R1 I'-4;
or a pharmaceutically acceptable salt thereof, and the variables are as
defined in the first
embodiment.
In a sixth embodiment, the application provides a compound according to
Structural
Formula I', I'-1, I'-2, I'-3, or I'-4, or a pharmaceutically acceptable salt
thereof, in
combination with a PARP inhibitor, wherein X4 is NH, and the remaining
variables are as
defined in the first embodiment.
In a seventh embodiment, the application provides a compound according to
Structural Formula I', I'-1, I'-2, I'-3, or I'-4, or a pharmaceutically
acceptable salt thereof, in
combination with a PARP inhibitor, wherein le is (C1¨C4)alkyl, -
(C4¨C6)cycloalkyl, ¨
(CH2)qheterocycly1 (wherein the heterocycyl is a monocyclic 4-6-membered
heterocyclic ring
optionally substituted with one methyl), or benzyl, and the remaining
variables are as defined
in the first and/or sixth embodiments. In one specific embodiment, le is
isopropyl, tert-butyl,
19

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
cyclobutyl, cyclopentyl, oxetanyl, benzyl, tetrahydro-2H-pyranyl, or . In
another
specific embodiment, le is isopropyl or oxetanyl.
In an eighth embodiment, the application provides a compound according to
Structural Formula I', I'-1, I'-2, I'-3, or I'-4, or a pharmaceutically
acceptable salt thereof, in
combination with a PARP inhibitor, wherein Rd is halogen, and m is 0 or 1, and
the
remaining variables are as defined in the first, sixth, and/or seventh
embodiments. In one
_Cyy (1X.y5 Clxly
(Rd)M I I
specific embodiment, is C0s, , or
(ros5N :
In a ninth embodiment, the application provides a compound according to
Structural
Formula I', I'-1, I'-2, I'-3, or I'-4, or a pharmaceutically acceptable salt
thereof, in
combination with a PARP inhibitor, wherein le is tert-butyl, and the remaining
variables are
as defined in the first, sixth, seventh, and/or eighth embodiments.
In a tenth embodiment, the application provides a compound, or a
pharmaceutically
acceptable salt thereof, in combination with a PARP inhibitor, wherein the
compound is
selected from the group consisting of:
0
0
S
IN 0
GNAHN H
H HN µ0
0
0
S
crNAHN SH
H HN \O
,and

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
N

)\..0 0
0
N__1L A 101 S
HN H
HN \O
In an eleventh embodiment, the application provides a compound represented by
Structural Formula II' in combination with a PARP inhibitor:
, N
R¨X
/ \ 0
0
A.J.L X5X6-0
HN
HN \O
R1 II';
or a pharmaceutically acceptable salt thereof, wherein:
the thiazole ring is optionally substituted with -F or ¨Cl;
X4 is NRa or 0;
X5 and X6 are each independently NRb or 0;
R1 is (Ci¨05)alkyl;
R4 is (C1¨C4)alkyl, -(C3¨C7)cycloalkyl, ¨(CH(Rc))q-heterocycyl (wherein the
heterocycyl is a monocyclic 3-7-membered heterocyclic ring optionally
substituted with one
or more occurences of methyl), ¨(CH(Rc))q-phenyl (wherein the phenyl ring is
optionally
substituted with one or more occurences of halogen, methoxy, halomethoxy,
methyl,
halomethyl, or cyano), or ¨(CH(Rc))q-2-pyridinyl (wherein the 2-pyridinyl ring
is optionally
substituted with one or more occurences of halogen, methoxy, halomethoxy,
methyl,
halomethyl, or cyano);
each of Ra, Rb, and RC is independently hydrogen or methyl;
n is 0, 1, or 2; and
q is 0 or 1.
In a twelveth embodiment, the application provides a compound represented by
Structural Formula II'-1 in combination with a PARP inhibitor:
0
R¨X
0 s
A AJL HN 0¨00
HN NO
R1 II'-!;
21

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
or a pharmaceutically acceptable salt thereof, and the variables are as
defined in the eleventh
embodiment.
In a thirteenth embodiment, the application provides a compound represented by

Structural Formula II'-2 in combination with a PARP inhibitor:
, N
/ 0
0
R4¨X4jLHN I
HN \O
R1 II'-2;
or a pharmaceutically acceptable salt thereof, and the variables are as
defined in the eleventh
embodiment.
In a fourteenth embodiment, the application provides a compound according to
Structural Formula II', II'-1 or II'-2, or a pharmaceutically acceptable salt
thereof, in
combination with a PARP inhibitor, wherein le is isopropyl, oxetanyl,
cyclobutyl, ¨CH2-2-
pyrrolidinyl, ¨CH2-N-methyl-2-pyrrolidinyl, ¨CH2-3-piperidinyl, ¨CH2-2-
pyrazinyl, ¨CH2-2-
pyrimidinyl, ¨CH(Rc)-phenyl, or ¨CH(Rc)-2-pyridinyl, and that the phenyl and 2-
pyridinyl
rings are each independently and optionally substituted with one or more
occurences of
halogen, and the remaining variables are as defined in the eleventh
embodiment. In one
specific embodiment, le is .1 110
F =
, or . In another specific embodiment, le is
F =
101
or
In a fifteenth embodiment, the application provides a compound according to
Structural Formula II', II'-1 or II'-2, or a pharmaceutically acceptable salt
thereof, in
combination with a PARP inhibitor, wherein X' is NH, and the remaining
variables are as
defined in the eleventh and/or fourteenth embodiments.
In a sixteenth embodiment, the application provides a compound according to
Structural Formula II', II'-1 or II'-2, or a pharmaceutically acceptable salt
thereof, in
22

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
combination with a PARP inhibitor, wherein It' is tert-butyl, and the
remaining variables are
as defined in the eleventh, fourteenth, and fifteenth embodiments.
In a seventeenth embodiment, the application provides a compound, or a
pharmaceutically acceptable salt thereof, in combination with a PARP
inhibitor, wherein the
compound is selected from the group consisting of:
O
0
NAHN H
HN
NAHN
and
, N
/ 0
0 S
-IN 0¨00
,C) H
HN \O
In a eighteenth embodiment, the application provides a compound, or a
pharmaceutically acceptable salt thereof, in combination with a PARP
inhibitor, wherein the
compound is:
0
I
I 4)
N S=0
HNI
Also included are the compounds disclosed in the Exemplification, both in the
pharmaceutically acceptable salt form and in the neutral form, in combination
with a PARP
inhibitor.
The term "pharmaceutically acceptable salt" refers to a pharmaceutical salt
that is,
within the scope of sound medical judgment, suitable for use in contact with
the tissues of
humans and lower animals without undue toxicity, irritation, and allergic
response, and is
commensurate with a reasonable benefit/risk ratio. Pharmaceutically-acceptable
salts are well
known in the art. For example, S. M. Berge et at. describes pharmacologically
acceptable
salts in I Pharm. Sc., 1977, 66, 1-19.
Included in the present application are pharmaceutically acceptable salts of
the
23

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
compounds disclosed herein. Compounds having basic groups can form
pharmaceutically
acceptable salts with pharmaceutically acceptable acid(s). Suitable
pharmaceutically
acceptable acid addition salts of the compounds described herein include salts
of inorganic
acids (such as hydrochloric acid, hydrobromic, phosphoric, metaphosphoric,
nitric, and
sulfuric acids) and of organic acids (such as acetic acid, benzenesulfonic,
benzoic,
ethanesulfonic, methanesulfonic, succinic, and trifluoroacetic acid acids).
Compounds of the
present application with acidic groups such as carboxylic acids can form
pharmaceutically
acceptable salts with pharmaceutically acceptable base(s). Suitable
pharmaceutically
acceptable basic salts include ammonium salts, alkali metal salts (such as
sodium and
potassium salts) and alkaline earth metal salts (such as magnesium and calcium
salts).
In one aspect, this application provides a combination of a RAD51 inhibitor
and a
PARP inhibitor. As used herein, a poby- ADP ribose poby-merase (PART) is a
member of a
family of proteins that is involved in a number of cellular processes, such as
DNA repair and
programmed cell death..A PARP inhibitor (PARPi) reduces the functioning of a
poly ADP
ribose polyrnerase. In particular embodiments, the PARP inhibitor is selected
from the group
consisting of veliparib, BIVIN-673, 4-iodo-3-nitrobenzamide, olaparib,
rucaparib, CEP 9722,
niraparib, talazoparib (BMN-673), pamiparib (BGB-290), iniparib (BSI-201,
5AR240550),
INO-1001, ABT-767, E7016 / GPI-21016, AZD2461, "12D2281, AIM-100, and 2X-121.
In some embodiments, the PARP inhibitor is selected from the group consisting
of
olaparib, veliparib, rucaparib, talazoparib, and niraparib. In some
embodiments, the PARP
inhibitor is olaparib. In some embodiments, the PARP inhibitor is veliparib.
In some
embodiments, the PARP inhibitor is rucaparib. In 'vet some embodiments, the
PART' inhibitor
is talazoparib. In some embodiments the PARP inhibitor is nira.parib.
Definitions
The term "synergistic" as used herein refers to a therapeutic combination
which is
more effective than the additive effects of the two or more single agents. A
determination of a
synergistic interaction between the compounds disclosed herein, and one or
more PARP
inhibitor may be based on the results obtained from the assays described
herein.
The combination therapy may provide "synergy" and prove "synergistic", i.e.,
the effect
achieved when the active ingredients used together is greater than the sum of
the effects that
results from using the compounds separately. A synergistic effect may be
attained when the
active ingredients are: (1) co-formulated and administered or delivered
simultaneously in a
24

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
combined, unit dosage formulation; (2) delivered by alternation as separate
formulations; or
(3) by some other regimen. When delivered in alternation therapy, a
synergistic effect may be
attained when the compounds are administered or delivered sequentially. In
some
embodiments, the compound of the present application is administered first. In
other
embodiments, the PARP inhibitor is administered first. In alternate
embodimnets, the
combination therapy is administered concomitantly as separate compositions. In
alternate
embodiments, the combination therapy is administered as a single composition.
Combination therapy in which two or more drugs are used together in some
dosing
regimen or administration form, typically has one or more goals of: (i)
reducing the
frequency at which acquired resistance arises by combining drugs with minimal
cross-
resistance, (ii) lowering the doses of drugs with non-overlapping toxicity and
similar
therapeutic profile so as to achieve efficacy with fewer side effects, i.e.,
increase therapeutic
index, (iii) sensitizing cells to the action of one drug through use of
another drug, such as
altering cell-cycle stage or growth properties, and (iv) achieving enhanced
potency by
exploiting additivity, or greater than additivity, effects in the biological
activity of two drugs.
As used herein, the term "combination", as applied to two or more compounds
and/or
agents, is intended to define material in which the two or more agents are
associated. The
terms "combined" and "combining" in this context are to be interpreted
accordingly.
The association of the two or more compounds/agents in a combination may be
physical or non-physical. Examples of physically associated combined
compounds/agents
include: compositions (e.g., unitary formulations) comprising the two or more
compounds/agents in admixture (for example within the same unit dose);
compositions
comprising material in which the two or more compounds/agents are
chemically/physicochemically linked (for example by crosslinking, molecular
agglomeration
or binding to a common vehicle moiety); compositions comprising material in
which the two
or more compounds/agents are chemically/physicochemically co-packaged (for
example,
disposed on or within lipid vesicles, particles (e.g. micro- or nanoparticles)
or emulsion
droplets); pharmaceutical kits, pharmaceutical packs or patient packs in which
the two or
more compounds/agents are co-packaged or co-presented (e.g. as part of an
array of unit
doses). Examples of non-physically associated combined compounds/agents
include: material
(e.g., a non-unitary formulation) comprising at least one of the two or more
compounds/agents together with instructions for the extemporaneous association
of the at
least one compound to form a physical association of the two or more
compounds/agents;

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
material (e.g., a non-unitary formulation) comprising at least one of the two
or more
compounds/agents together with instructions for combination therapy with the
two or more
compounds/agents; material comprising at least one of the two or more
compounds/agents
together with instructions for administration to a patient population in which
the other(s) of
the two or more compounds/agents have been (or are being) administered;
material
comprising at least one of the two or more compounds/agents in an amount or in
a form
which is specifically adapted for use in combination with the other(s) of the
two or more
compounds/agents.
As used herein, the term "combination therapy" is intended to define therapies
which
comprise the use of a combination of two or more compounds/agents (as defined
above).
Thus, references to "combination therapy", "combinations" and the use of
compounds/agents
"in combination" in this application may refer to compounds/agents that are
administered as
part of the same overall treatment regimen. As such, the dosage regimine of
each of the two
or more compounds/agents may differ: each may be administered at the same time
or at
different times. It will therefore be appreciated that the compounds/agents of

the combination may be administered sequentially (e.g. before or after) or
simultaneously,
either in the same pharmaceutical formulation (i.e. together), or in different
pharmaceutical
formulations (i.e. separately). Simultaneously in the same formulation is as a
unitary
formulation whereas simultaneously in different pharmaceutical formulations is
non-unitary.
The dosage of each of the two or more compounds/agents in a combination
therapy may also
differ with respect to the route of administration.
As used herein, the term "pharmaceutical kit" defines an array of one or more
unit
doses of a pharmaceutical composition together with dosing means (e.g.
measuring device)
and/or delivery means (e.g. inhaler or syringe), optionally all contained
within common outer
packaging. In pharmaceutical kits comprising a combination of two or more
compounds/agents, the individual compounds/agents may be unitary or non-
unitary
formulations. The unit dose(s) may be contained within a blister pack. The
pharmaceutical kit
may optionally further comprise instructions for use.
As used herein, the term "pharmaceutical pack" defines an array of one or more
unit
doses of a pharmaceutical composition, optionally contained within common
outer
packaging. In pharmaceutical packs comprising a combination of two or more
compounds/agents, the individual compounds/agents may be unitary or non-
unitary
formulations. The unit dose(s) may be contained within a blister pack. The
pharmaceutical
26

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
pack may optionally further comprise instructions for use.
In some embodiments, "temporal proximity" means that administration of one
therapeutic agent occurs within a time period before or after the
administration of another
therapeutic agent, such that the therapeutic effect of the one therapeutic
agent overlaps with
the therapeutic effect of the another therapeutic agent. In some embodiments,
the therapeutic
effect of the one therapeutic agent completely overlaps with the therapeutic
effect of the other
therapeutic agent. In some embodiments, "temporal proximity" means that
administration of
one therapeutic agent occurs within a time period before or after the
administration of another
therapeutic agent, such that there is a synergistic effect between the one
therapeutic agent and
the another therapeutic agent. "Temporal proximity" may vary according to
various factors,
including but not limited to, the age, gender, weight, genetic background,
medical condition,
disease history, and treatment history of the subject to which the therapeutic
agents are to be
administered; the disease or condition to be treated or ameliorated; the
therapeutic outcome to
be achieved; the dosage, dosing frequency, and dosing duration of the
therapeutic agents; the
pharmacokinetics and pharmacodynamics of the therapeutic agents; and the
route(s) through
which the therapeutic agents are administered. In some embodiments, "temporal
proximity"
means within 15 minutes, within 30 minutes, within an hour, within two hours,
within four
hours, within six hours, within eight hours, within 12 hours, within 18 hours,
within 24 hours,
within 36 hours, within 2 days, within 3 days, within 4 days, within 5 days,
within 6 days,
within a week, within 2 weeks, within 3 weeks, within 4 weeks, with 6 weeks,
or within 8
weeks. In some embodiments, multiple administration of one therapeutic agent
can occur in
temporal proximity to a single administration of another therapeutic agent. In
some
embodiments, temporal proximity may change during a treatment cycle or within
a dosing
regimen.
The term "halo" as used herein means halogen and includes fluor , chloro,
bromo and
iodo.
The term "alkyl" used alone or as part of a larger moiety, such as "alkoxy" or

"haloalkyl" and the like, means saturated aliphatic straight-chain or branched
monovalent
hydrocarbon radical. Unless otherwise specified, an alkyl group typically has
1-5 carbon
atoms, i.e. (Ci-05)alkyl. As used herein, a "(Ci-05)alkyl" group means a
radical having from
1 to 5 carbon atoms in a linear or branched arrangement. Examples include
methyl, ethyl, n-
propyl, iso-propyl, and the like.
27

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
The term "alkoxy" means an alkyl radical attached through an oxygen linking
atom,
represented by ¨0-alkyl. For example, "(C1-C4)alkoxy" includes methoxy,
ethoxy, propoxy,
and butoxy.
The terms "haloalkyl" and "haloalkoxy" means alkyl or alkoxy, as the case may
be,
substituted with one or more halogen atoms.
An "alkylene group" is a saturated aliphatic branched or straight-chain
divalent
hydrocarbon radical. Unless otherwise specified, an alkylene group typically
has 2-6 carbon
atoms, e.g. (C2-C6)alkylene.
The term "alkenyl" means branched or straight-chain monovalent hydrocarbon
radical
containing at least one double bond. Alkenyl may be mono or polyunsaturated,
and may exist
in the E or Z configuration. Unless otherwise specified, an alkenyl group
typically has 2-6
carbon atoms, i.e., (C2-C6)alkenyl. For example, "(C2-C4)alkenyl" means a
radical having
from 2-4 carbon atoms in a linear or branched arrangement.
The term "cycloalkyl" means a monocyclic saturated hydrocarbon ring system.
For
example, a C3-C6 cycloalkyl includes cyclopropyl, cyclobutyl, cyclopentyl and
cyclohexyl.
Unless otherwise described, a "cycloalkyl" has from three to seven ring carbon
atoms.
A bridged cycloalkyl means a bicyclic non-aromatic hydrocarbon ring system in
which the two rings share at least three adjacent ring carbon atoms. A bridged
cycloalkyl
typically has 6-12 ring carbon atoms. Examples include, but are not limited
to,
bicyclo[2.1.1]hexyl, bicyclo[2.2.1]heptyl, bicyclo[2.2.2]octyl,
bicyclo[3.2.1]octyl,
bicyclo[4.3.1]decyl, bicyclo[3.3.1]nonyl, bornyl, bornenyl, norbornyl,
norbornenyl, 6,6-
dimethylbicyclo [3.1.1]heptyl, tricyclobutyl, and adamantyl.
The terms "heterocyclyl", "heterocyclic ring", and "heterocyclic group", are
used
interchangeably herein, and means a saturated or unsaturated non-aromatic 4-10
membered
ring radical containing from 1 to 4 ring heteroatoms, which may be the same or
different,
selected from N, 0, or S. It can be monocyclic, bicyclic or tricyclic (e.g., a
fused or bridged
bicyclic or tricyclic ring). Examples of include, but are not limited to,
azetidinyl,
morpholinyl, thiomorpholinyl, pyrrolidinonyl, pyrrolidinyl, piperidinyl,
piperazinyl,
hydantoinyl, valerolactamyl, oxiranyl, oxetanyl, dihydroimidazole,
dihydrofuranyl,
dihydropyranyl, dihydropyridinyl, dihydropyrimidinyl, dihydrothienyl,
dihydrothiophenyl,
dihydrothiopyranyl, tetrahydroimidazole, tetrahydrofuranyl, tetrahydropyranyl,

tetrahydrothienyl, tetrahydropyridinyl, tetrahydropyrimidinyl,
tetrahydrothiophenyl, and
tetrahydrothiopyranyl. A heterocyclic ring optionally contains one or more
double bonds
28

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
and/or is optionally fused with one or more aromatic rings (for example,
tetrahydronaphthyridine, indolinone, dihydropyrrolotriazole,
imidazopyrimidine,
quinolinone, dioxaspirodecane).
Examples of 3-7 membered monocyclic heterocyclic ring include, but are not
limited
to, azetidinyl, morpholinyl, thiomorpholinyl, pyrrolidinonyl, pyrrolidinyl,
piperidinyl,
piperazinyl, hydantoinyl, valerolactamyl, oxiranyl, oxetanyl,
dihydroimidazole,
dihydrofuranyl, dihydropyranyl, dihydropyridinyl, dihydropyrimidinyl,
dihydrothienyl,
dihydrothiophenyl, dihydrothiopyranyl, tetrahydroimidazole, tetrahydrofuranyl,

tetrahydropyranyl, tetrahydrothienyl, tetrahydropyridinyl,
tetrahydropyrimidinyl,
tetrahydrothiophenyl, and tetrahydrothiopyranyl.
A bridged heterocyclyl means a bicyclic non-aromatic ring system containing
from 1
to 4 ring heteroatoms in which the two rings share at least three adjacent
ring atoms. A
bridged heterocyclyl typically has 6-12 ring atoms. Examples include, but are
not limited to,
azanorbornyl, quinuclidinyl, isoquinuclidinyl, tropanyl,
azabicyclo[3.2.1]octanyl,
azabicyclo[2.2. 1 ]heptany 1, 2-azabicyclo[3 .2. 1 ]octanyl, azabicyclo[3 .2.
1 ]octanyl,
azabicyclo[3.2.2]nonanyl, azabicyclo[3.3.0]nonanyl, and azabicyclo
[3.3.1]nonanyl.
The terms "heteroaryl", "heteroaromatic", "heteroaryl ring", "heteroaryl
group",
"heteroaromatic ring", and "heteroaromatic group", are used interchangeably
herein.
"Heteroaryl" when used alone or as part of a larger moiety as in
"heteroaralkyl" or
"heteroarylalkoxy", refers to aromatic ring groups having five to ten ring
atoms selected
from carbon and at least one (typically 1 to 4, more typically 1 or 2)
heteroatoms (e.g.,
oxygen, nitrogen or sulfur). "Heteroaryl" includes monocyclic rings and
polycyclic rings in
which a monocyclic heteroaromatic ring is fused to one or more other aromatic
or
heteroaromatic rings. "Heteroaryl" includes monocyclic and bicyclic ring
systems.
"Monocyclic 5-6 membered heteroaromatic ring (or heteroaryl)" means a
monocyclic
heteroaromatic ring having five or six ring atoms selected from carbon and at
least one
(typically 1 to 3, more typically 1 or 2) heteroatoms (e.g., oxygen, nitrogen
or sulfur).
Examples of monocyclic 5-6 membered heteroaromatic ring groups include furanyl
(e.g., 2-
furanyl, 3-furanyl), imidazolyl (e.g., N-imidazolyl, 2-imidazolyl, 4-
imidazolyl, 5-imidazoly1),
isoxazolyl (e.g., 3-isoxazolyl, 4-isoxazolyl, 5-isoxazoly1), oxadiazolyl
(e.g., 2-oxadiazolyl, 5-
oxadiazolyl), oxazolyl (e.g., 2-oxazolyl, 4-oxazolyl, 5-oxazoly1), pyrazolyl
(e.g., 3-pyrazolyl,
4-pyrazoly1), pyrrolyl (e.g., 1-pyrrolyl, 2-pyrrolyl, 3-pyrroly1), pyridyl
(e.g., 2-pyridyl, 3-
pyridyl, 4-pyridy1), pyrimidinyl (e.g., 2-pyrimidinyl, 4-pyrimidinyl, 5-
pyrimidinyl),
29

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
pyridazinyl (e.g., 3-pyridazinyl), thiazolyl (e.g., 2-thiazolyl, 4-thiazolyl,
5-thiazoly1),
isothiazolyl, triazolyl (e.g., 2-triazolyl, 5-triazoly1), tetrazolyl (e.g.,
tetrazolyl), and thienyl
(e.g., 2-thienyl, 3-thieny1).
If a group is described as being "substituted," a non-hydrogen substituent
replaces a
hydrogen on a carbon or nitrogen of the substituent. Thus, for example, a
substituted alkyl is
an alkyl wherein at least one non-hydrogen substituent is in the place of a
hydrogen
substituent on the alkyl substituent. To illustrate, monofluoroalkyl is alkyl
substituted with a
fluoro substituent, and difluoroalkyl is alkyl substituted with two fluoro
substituents. It
should be recognized that if there is more than one substitution on a
substituent, each non-
hydrogen substituent can be identical or different (unless otherwise stated).
As used herein,
many moieties (e.g., alkyl, cycloalkyl, or a heterocyclic ring) are referred
to as being either
"substituted" or "optionally substituted". When a moiety is modified by one of
these terms,
unless otherwise noted, it denotes that any portion of the moiety that is
known to one skilled
in the art as being available for substitution can be substituted, which
includes one or more
substituents. If more than one substituent is present, then each substituent
is independently
selected. Such means for substitution are well-known in the art and/or taught
by the instant
application. The optional substituents can be any substituents that are
suitable to attach to the
moiety. A person of ordinary skill in the art will recognize that the
compounds and
definitions provided do not include impermissible substituent patterns (e.g.,
methyl
substituted with 5 different groups, and the like). Such impermissible
substitution patterns are
clearly recognized by a person of ordinary skill in the art. When a group is
described as being
optionally substituted by "one or more" substituents, it denotes that the
group is optionally
substituted by one, two, three, four, five or six substituents. In one
embodiment, a group is
optionally substituted by 1-3 substituents. In one embodiment, a group is
optionally
substituted by 1-2 substituents. In one embodiment, a group is optionally
substituted by one
substituent.
Suitable substituents are those which do not have a significant adverse effect
on the
ability of the compound to inhibit RAD51. Where suitable substituents are not
specifically
enumerated, exemplary substituents include, but are not limited to, halo, -CN,
alkyl, alkoxy,
halomethyl, halomethoxy, (Ci-05)alkyl, halo(Ci-05)alkyl, (Ci-05)alkoxy, -NO2,
-
NRa'Rb', -S(0)Ra', -NRa5(0)Rb', -S(0)1NRa'Rb', -C(=0)0Ra', -0C(=0)0Ra', -
C(=5)0Ra', -
0(C=S)Ra', -C(=0)NRa'Rb', -N1a'C(=0)Rb', -C(=S)NRale', -NRa'C(=S)Rb',

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
4Ra'(C=0)0Rb', - 0 (C= 0)1\TRa' Rb - NRa'(C= S)ORb - 0 (C = S)1\TRale',
41a'(C=0)1\TRale',
-NRa'(C= S)I\TRa'Rb', -C(¨S)Ra', -C(-0)Ra', (C3-C6)cycloalkyl, monocyclic
heteroaryl and
phenyl, wherein the (C3-C6)cycloalkyl, monocyclic heteroaryl and phenyl
substituents are
optionally and independently substituted with ¨CH3, halomethyl, halo, methoxy
or
halomethoxy. Each Ra' and each Rb' are independently selected from ¨H and
(Ci¨05)alkyl,
wherein the (Ci¨05)alkyl group represented by Ra' or Rb' is optionally
substituted with
hydroxyl or (C1¨C3)alkoxy; Rc' is ¨H, halo(Ci¨05)alkyl or (Ci¨05)alkyl,
wherein the
(Ci¨05)alkyl group represented by RC is optionally substituted with hydroxyl
or
(C1¨C3)alkoxy; and i is 0, 1, or 2. =0 is also a suitable substituent for
alkyl, cycloalkyl, and a
heterocyclic ring.
Compounds having one or more chiral centers can exist in various
stereoisomeric
forms. Stereoisomers are compounds that differ only in their spatial
arrangement.
Stereoisomers include all diastereomeric, enantiomeric, and epimeric forms as
well as
racemates and mixtures thereof.
The term "geometric isomer" refers to cyclic compounds having at least two
substituents, wherein the two substituents are both on the same side of the
ring (cis) or
wherein the substituents are each on opposite sides of the ring (trans). When
a disclosed
compound is named or depicted by structure without indicating stereochemistry,
it is
understood that the name or the structure encompasses one or more of the
possible
stereoisomers, or geometric isomers, or a mixture of the encompassed
stereoisomers or
geometric isomers.
When a geometric isomer is depicted by name or structure, it is to be
understood that
the named or depicted isomer exists to a greater degree than another isomer,
that is that the
geometric isomeric purity of the named or depicted geometric isomer is greater
than 50%,
such as at least 60%, 70%, 80%, 90%, 99%, or 99.9% pure by weight. Geometric
isomeric
purity is determined by dividing the weight of the named or depicted geometric
isomer in the
mixture by the total weight of all of the geometric isomers in the mixture.
Racemic mixture means 50% of one enantiomer and 50% of is corresponding
enantiomer. When a compound with one chiral center is named or depicted
without indicating
the stereochemistry of the chiral center, it is understood that the name or
structure
encompasses both possible enantiomeric forms (e.g., both enantiomerically-
pure,
enantiomerically-enriched or racemic ) of the compound. When a compound with
two or
more chiral centers is named or depicted without indicating the
stereochemistry of the chiral
31

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
centers, it is understood that the name or structure encompasses all possible
diastereomeric
forms (e.g., diastereomerically pure, diastereomerically enriched and
equimolar mixtures of
one or more diastereomers (e.g., racemic mixtures) of the compound.
Enantiomeric and diastereomeric mixtures can be resolved into their component
enantiomers or stereoisomers by well-known methods, such as chiral-phase gas
chromatography, chiral-phase high performance liquid chromatography,
crystallizing the
compound as a chiral salt complex, or crystallizing the compound in a chiral
solvent.
Enantiomers and diastereomers also can be obtained from diastereomerically- or

enantiomerically-pure intermediates, reagents, and catalysts by well-known
asymmetric
synthetic methods.
When a compound is designated by a name or structure that indicates a single
enantiomer, unless indicated otherwise, the compound is at least 60%, 70%,
80%, 90%, 99%
or 99.9% optically pure (also referred to as "enantiomerically pure"). Optical
purity is the
weight in the mixture of the named or depicted enantiomer divided by the total
weight in the
mixture of both enantiomers.
When the stereochemistry of a disclosed compound is named or depicted by
structure,
and the named or depicted structure encompasses more than one stereoisomer
(e.g., as in a
diastereomeric pair), it is to be understood that one of the encompassed
stereoisomers or any
mixture of the encompassed stereoisomers is included. It is to be further
understood that the
stereoisomeric purity of the named or depicted stereoisomers at least 60%,
70%, 80%, 90%,
99% or 99.9% by weight. The stereoisomeric purity in this case is determined
by dividing the
total weight in the mixture of the stereoisomers encompassed by the name or
structure by the
total weight in the mixture of all of the stereoisomers.
Pharmaceutical Compositions
The compounds disclosed therein are RADS I inhibitors. The pharmaceutical
composition of the present application comprises one or more RAD51 inhibitors,
or a
pharmaceutically acceptable salt thereof, and one or more PARP inhibitors, or
a
pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable
carrier or diluent.
"Pharmaceutically acceptable carrier" and "pharmaceutically acceptable
diluent" refer
to a substance that aids the formulation and/or administration of an active
agent to and/or
absorption by a subject and can be included in the compositions of the present
application
without causing a significant adverse toxicological effect on the subject. Non-
limiting
examples of pharmaceutically acceptable carriers and/or diluents include
water, NaCl, normal
32

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
saline solutions, lactated Ringer's, normal sucrose, normal glucose, binders,
fillers,
disintegrants, lubricants, coatings, sweeteners, flavors, salt solutions (such
as Ringer's
solution), alcohols, oils, gelatins, carbohydrates such as lactose, amylose or
starch, fatty acid
esters, hydroxymethycellulose, polyvinyl pyrrolidine, and colors, and the
like. Such
preparations can be sterilized and, if desired, mixed with auxiliary agents
such as lubricants,
preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing
osmotic pressure,
buffers, coloring, and/or aromatic substances and the like that do not
deleteriously react with
or interfere with the activity of the compounds provided herein and PARP
inhibitors. One of
ordinary skill in the art will recognize that other pharmaceutical excipients
are suitable for
use with disclosed compounds and PARP inhibitors.
The pharmaceutical compositions of the present application optionally include
one or
more pharmaceutically acceptable carriers and/or diluents therefor, such as
lactose, starch,
cellulose and dextrose. Other excipients, such as flavoring agents;
sweeteners; and
preservatives, such as methyl, ethyl, propyl and butyl parabens, can also be
included. More
complete listings of suitable excipients can be found in the Handbook of
Pharmaceutical
Excipients (5th Ed., Pharmaceutical Press (2005)). A person skilled in the art
would know
how to prepare formulations suitable for various types of administration
routes. Conventional
procedures and ingredients for the selection and preparation of suitable
formulations are
described, for example, in Remington's Pharmaceutical Sciences (2003 - 20th
edition) and in
The United States Pharmacopeia: The National Formulary (USP 24 NF19) published
in 1999.
The carriers, diluents and/or excipients are "acceptable" in the sense of
being compatible with
the other ingredients of the pharmaceutical composition and not deleterious to
the recipient
thereof
A "pharmaceutical composition" contains the compound of the present
application
and a PARP inhibitor in a form suitable for administration to a subject. In
one embodiment,
the pharmaceutical composition is in bulk or in unit dosage form. The unit
dosage form is any
of a variety of forms, including, for example, a capsule, an IV bag, a tablet,
a single pump on
an aerosol inhaler or a vial. The quantity of active ingredient (e.g., a
formulation of the
disclosed compound or a pharmaceutically acceptable salt thereof and a
formulation of a
PARP inhibitor or a a pharmaceutically acceptable salt thereof) in a unit dose
of composition
is an effective amount and is varied according to the particular treatment
involved. One
skilled in the art will appreciate that it is sometimes necessary to make
routine variations to
the dosage depending on the age and condition of the patient. The dosage will
also depend on
33

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
the route of administration. A variety of routes are contemplated, including
oral, pulmonary,
rectal, parenteral, transdermal, subcutaneous, intravenous, intramuscular,
intraperitoneal,
inhalational, buccal, sublingual, intrapleural, intrathecal, intranasal, and
the like. Dosage
forms for the topical or transdermal administration of a compound of this
application and a
PARP inhibitor include powders, sprays, ointments, pastes, creams, lotions,
gels, solutions,
patches and inhalants. In one embodiment, the active compound and the PARP
inhibitor is
mixed under sterile conditions with a pharmaceutically acceptable carrier, and
with any
preservatives, buffers or propellants that are required.
As used herein, the phrase "pharmaceutically acceptable" refers to those
compounds,
materials, compositions, carriers, and/or dosage forms which are, within the
scope of sound
medical judgment, suitable for use in contact with the tissues of human beings
and animals
without excessive toxicity, irritation, allergic response, or other problem or
complication,
commensurate with a reasonable benefit/risk ratio.
"Pharmaceutically acceptable carrier" and "pharmaceutically acceptable
diluent" refer
to a substance that aids the formulation and/or administration of an active
agent to and/or
absorption by a subject and can be included in the compositions of the present
application
without causing a significant adverse toxicological effect on the subject. Non-
limiting
examples of pharmaceutically acceptable carriers and/or diluents include
water, NaCl, normal
saline solutions, lactated Ringer's, normal sucrose, normal glucose, binders,
fillers,
disintegrants, lubricants, coatings, sweeteners, flavors, salt solutions (such
as Ringer's
solution), alcohols, oils, gelatins, carbohydrates such as lactose, amylose or
starch, fatty acid
esters, hydroxymethycellulose, polyvinyl pyrrolidine, and colors, and the
like. Such
preparations can be sterilized and, if desired, mixed with auxiliary agents
such as lubricants,
preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing
osmotic pressure,
buffers, coloring, and/or aromatic substances and the like that do not
deleteriously react with
or interfere with the activity of the compounds and PARP inhibitors provided
herein. One of
ordinary skill in the art will recognize that other pharmaceutical excipients
are suitable for
use with disclosed compounds and PARP inhibitors.
The term "carrier", as used in this application, encompasses carriers,
excipients, and
diluents and means a material, composition or vehicle, such as a liquid or
solid filler, diluent,
excipient, solvent or encapsulating material, involved in carrying or
transporting a
pharmaceutical agent from one organ, or portion of the body, to another organ,
or portion of
the body of a subject.
34

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
The pharmaceutical compositions of the present application optionally include
one or
more pharmaceutically acceptable carriers and/or diluents therefor, such as
lactose, starch,
cellulose and dextrose. Other excipients, such as flavoring agents;
sweeteners; and
preservatives, such as methyl, ethyl, propyl and butyl parabens, can also be
included. More
complete listings of suitable excipients can be found in the Handbook of
Pharmaceutical
Excipients (5th Ed., Pharmaceutical Press (2005)). A person skilled in the art
would know
how to prepare formulations suitable for various types of administration
routes. Conventional
procedures and ingredients for the selection and preparation of suitable
formulations are
described, for example, in Remington's Pharmaceutical Sciences (2003 - 20th
edition) and in
The United States Pharmacopeia: The National Formulary (USP 24 NF19) published
in 1999.
The carriers, diluents and/or excipients are "acceptable" in the sense of
being compatible with
the other ingredients of the pharmaceutical composition and not deleterious to
the recipient
thereof
Pharmaceutical compositions of the application are formulated to be compatible
with
its intended route of administration. Examples of routes of administration
include parenteral,
e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation),
transdermal (topical), and
transmucosal administration. Solutions or suspensions used for parenteral,
intradermal, or
subcutaneous application can include the following components: a sterile
diluent such as
water for injection, saline solution, fixed oils, polyethylene glycols,
glycerine, propylene
glycol or other synthetic solvents; antibacterial agents such as benzyl
alcohol or methyl
parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating
agents such as
ethylenediaminetetraacetic acid; buffers such as acetates, citrates or
phosphates, and agents
for the adjustment of tonicity such as sodium chloride or dextrose. The pH can
be adjusted
with acids or bases, such as hydrochloric acid or sodium hydroxide. The
parenteral
preparation can be enclosed in ampoules, disposable syringes or multiple dose
vials made of
glass or plastic.
A compound or pharmaceutical composition can be administered to a subject in
many
of the well-known methods currently used for chemotherapeutic treatment. For
example, for
treatment of cancers, a compound may be injected directly into tumors,
injected into the
blood stream or body cavities or taken orally or applied through the skin with
patches. The
dose chosen should be sufficient to constitute effective treatment but not as
high as to cause
unacceptable side effects. The state of the disease condition (e.g., cancer,
precancer, and the

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
like) and the health of the patient should preferably be closely monitored
during and for a
reasonable period after treatment.
The term "therapeutically effective amount", as used herein, refers to an
amount of a
pharmaceutical agent to treat, ameliorate, or prevent an identified disease or
condition, or to
exhibit a detectable therapeutic or inhibitory effect. The effect can be
detected by any assay
method known in the art. The precise effective amount for a subject will
depend upon the
subject's body weight, size, and health; the nature and extent of the
condition; and the
therapeutic or combination of therapeutics selected for administration.
Therapeutically
effective amounts for a given situation can be determined by routine
experimentation that is
within the skill and judgment of the clinician.
For any compound, the therapeutically effective amount can be estimated
initially
either in cell culture assays, e.g., of neoplastic cells, or in animal models,
usually rats, mice,
rabbits, dogs, or pigs. The animal model may also be used to determine the
appropriate
concentration range and route of administration. Such information can then be
used to
determine useful doses and routes for administration in humans.
Therapeutic/prophylactic
efficacy and toxicity may be determined by standard pharmaceutical procedures
in cell
cultures or experimental animals, e.g., ED5o (the dose therapeutically
effective in 50% of the
population) and LD5o (the dose lethal to 50% of the population). The dose
ratio between toxic
and therapeutic effects is the therapeutic index, and it can be expressed as
the ratio,
LD5o/ED5o. Pharmaceutical compositions that exhibit large therapeutic indices
are preferred.
The dosage may vary within this range depending upon the dosage form employed,

sensitivity of the patient, and the route of administration.
Dosage and administration are adjusted to provide sufficient levels of the
active
agent(s) or to maintain the desired effect. Factors which may be taken into
account include
the severity of the disease state, general health of the subject, age, weight,
and gender of the
subject, diet, time and frequency of administration, drug combination(s),
reaction
sensitivities, and tolerance/response to therapy. Long-acting pharmaceutical
compositions
may be administered every 3 to 4 days, every week, or once every two weeks
depending on
half-life and clearance rate of the particular formulation.
The pharmaceutical compositions containing active compound of the present
application or PARP inhibitors may be manufactured in a manner that is
generally known,
e.g., by means of conventional mixing, dissolving, granulating, dragee-making,
levigating,
emulsifying, encapsulating, entrapping, or lyophilizing processes.
Pharmaceutical
36

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
compositions may be formulated in a conventional manner using one or more
pharmaceutically acceptable carriers comprising excipients and/or auxiliaries
that facilitate
processing of the active compound or PARP inhibitors into preparations that
can be used
pharmaceutically. Of course, the appropriate formulation is dependent upon the
route of
administration chosen.
Pharmaceutical compositions suitable for injectable use include sterile
aqueous
solutions (where water soluble) or dispersions and sterile powders for the
extemporaneous
preparation of sterile injectable solutions or dispersion. For intravenous
administration,
suitable carriers include physiological saline, bacteriostatic water,
Cremophor ELTM (BASF,
Parsippany, N.J.) or phosphate buffered saline (PBS). In all cases, the
composition must be
sterile and should be fluid to the extent that easy syringeability exists. It
must be stable under
the conditions of manufacture and storage and must be preserved against the
contaminating
action of microorganisms such as bacteria and fungi. The carrier can be a
solvent or
dispersion medium containing, for example, water, ethanol, polyol (for
example, glycerol,
propylene glycol, and liquid polyethylene glycol, and the like), and suitable
mixtures thereof.
The proper fluidity can be maintained, for example, by the use of a coating
such as lecithin,
by the maintenance of the required particle size in the case of dispersion and
by the use of
surfactants. Prevention of the action of microorganisms can be achieved by
various
antibacterial and antifungal agents, for example, parabens, chlorobutanol,
phenol, ascorbic
acid, thimerosal, and the like. In many cases, it will be preferable to
include isotonic agents,
for example, sugars, polyalcohols such as mannitol, sorbitol, sodium chloride
in the
composition. Prolonged absorption of the injectable compositions can be
brought about by
including in the composition an agent which delays absorption, for example,
aluminum
monostearate and gelatin.
Sterile injectable solutions can be prepared by incorporating the active
compound in
the required amount in an appropriate solvent with one or a combination of
ingredients
enumerated above, as required, followed by filtered sterilization. Generally,
dispersions are
prepared by incorporating the active compound into a sterile vehicle that
contains a basic
dispersion medium and the required other ingredients from those enumerated
above. In the
case of sterile powders for the preparation of sterile injectable solutions,
methods of
preparation are vacuum drying and freeze-drying that yields a powder of the
active ingredient
plus any additional desired ingredient from a previously sterile-filtered
solution thereof.
37

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
Oral compositions generally include an inert diluent or an edible
pharmaceutically
acceptable carrier. They can be enclosed in gelatin capsules or compressed
into tablets. For
the purpose of oral therapeutic administration, the active compound can be
incorporated with
excipients and used in the form of tablets, troches, or capsules. Oral
compositions can also be
prepared using a fluid carrier for use as a mouthwash, wherein the compound in
the fluid
carrier is applied orally and swished and expectorated or swallowed.
Pharmaceutically
compatible binding agents, and/or adjuvant materials can be included as part
of the
composition. The tablets, pills, capsules, troches and the like can contain
any of the following
ingredients, or compounds of a similar nature: a binder such as
microcrystalline cellulose,
gum tragacanth or gelatin; an excipient such as starch or lactose, a
disintegrating agent such
as alginic acid, Primogel, or corn starch; a lubricant such as magnesium
stearate or Sterotes; a
glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose
or saccharin; or a
flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
For administration by inhalation, the compound is delivered in the form of an
aerosol
spray from pressured container or dispenser, which contains a suitable
propellant, e.g., a gas
such as carbon dioxide, or a nebulizer.
Systemic administration can also be by transmucosal or transdermal means. For
transmucosal or transdermal administration, penetrants appropriate to the
barrier to be
permeated are used in the formulation. Such penetrants are generally known in
the art, and
include, for example, for transmucosal administration, detergents, bile salts,
and fusidic acid
derivatives. Transmucosal administration can be accomplished through the use
of nasal
sprays or suppositories. For transdermal administration, the active compound
is formulated
into ointments, salves, gels, or creams as generally known in the art.
The active compound can be prepared with pharmaceutically acceptable carriers
that
will protect the compound against rapid elimination from the body, such as a
controlled
release formulation, including implants and microencapsulated delivery
systems.
Biodegradable, biocompatible polymers can be used, such as ethylene vinyl
acetate,
polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic
acid. Methods for
preparation of such formulations will be apparent to those skilled in the art.
The materials can
also be obtained commercially from Alza Corporation and Nova Pharmaceuticals,
Inc.
Liposomal suspensions (including liposomes targeted to infected cells with
monoclonal
antibodies to viral antigens) can also be used as pharmaceutically acceptable
carriers. These
38

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
can be prepared according to methods known to those skilled in the art, for
example, as
described in U.S. Pat. No. 4,522,811.
It is especially advantageous to formulate oral or parenteral compositions in
dosage
unit form for ease of administration and uniformity of dosage. Dosage unit
form as used
herein refers to physically discrete units suited as unitary dosages for the
subject to be
treated; each unit containing a predetermined quantity of active compound
calculated to
produce the desired therapeutic effect in association with the required
pharmaceutical carrier.
The specification for the dosage unit forms of the application are dictated by
and directly
dependent on the unique characteristics of the active compound and the
particular therapeutic
effect to be achieved.
In therapeutic applications, the dosages of the pharmaceutical compositions
used in
accordance with the application vary depending on the agent, the age, weight,
and clinical
condition of the recipient patient, and the experience and judgment of the
clinician or
practitioner administering the therapy, among other factors affecting the
selected dosage.
Generally, the dose should be sufficient to result in slowing, and preferably
regressing, the
growth of the tumors and also preferably causing complete regression of the
cancer. Dosages
can range from about 0.01 mg/kg per day to about 5000 mg/kg per day. An
effective amount
of a pharmaceutical agent is that which provides an objectively identifiable
improvement as
noted by the clinician or other qualified observer. For example, regression of
a tumor in a
subject may be measured with reference to the diameter of a tumor. Decrease in
the diameter
of a tumor indicates regression. Regression is also indicated by failure of
tumors to reoccur
after treatment has stopped. As used herein, the term "dosage effective
manner" refers to
amount of an active compound to produce the desired biological effect in a
subject or cell.
The pharmaceutical compositions can be included in a container, pack, or
dispenser
together with instructions for administration.
The dosage regimen utilizing the compound is selected in accordance with a
variety
of factors including type, species, age, weight, sex and medical condition of
the patient; the
severity of the condition to be treated; the route of administration; the
renal and hepatic
function of the patient; and the particular compound or pharmaceutically
acceptable salt or
solvate thereof employed. An ordinarily skilled physician or veterinarian can
readily
determine and prescribe the effective amount of the drug required to prevent,
counter or
arrest the progress of the condition.
39

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
Techniques for formulation and administration of the disclosed compound of the

application and PARP inhibitors and combinations thereof can be found in
Remington: the
Science and Practice of Pharmacy, 19th edition, Mack Publishing Co., Easton,
PA (1995). In
an embodiment, the compound described herein, and the pharmaceutically
acceptable salts or
solvates thereof, and a PARP inhibitor, or a pharmaceutically acceptable salt
thereof, or a
pharmaceutical composition thereof, are used in pharmaceutical preparations in
combination
with a pharmaceutically acceptable carrier or diluent. Suitable
pharmaceutically acceptable
carriers include inert solid fillers or diluents and sterile aqueous or
organic solutions. The
compound or pharmaceutically acceptable salts thereof and the PARP inhibitor
or a
pharmaceutically acceptable salt thereof will be present in such
pharmaceutical compositions
in amounts sufficient to provide the desired dosage amount in the range
described herein.
All percentages and ratios used herein, unless otherwise indicated, are by
weight.
Other features and advantages of the present application are apparent from the
different
examples. The provided examples illustrate different components and
methodology useful in
practicing the present application. The examples do not limit the claimed
application. Based
on the present application the skilled artisan can identify and employ other
components and
methodology useful for practicing the present application.
Methods of Using the Combinations
In some embodiments, the present application provides a method of treating a
disease
or disorder, wherein the method comprises administering to a subject in need
thereof a
therapeutically effective amount of a compound described herein.
In some embodiments, the present application provides a method of treating a
disease
or disorder, wherein the method comprises administering to a subject in need
thereof a
composition comprising a RAD51 inhibitor, or a pharmaceutically acceptable
salt thereof,
and a PARP inhibitor, or a pharmaceutically acceptable salt thereof.
In some embodiments, the present application provides a method of treating a
disease
or disorder, wherein the method comprises administering to a subject in need
thereof a
combination of a compound of the present disclosure, or a pharmaceutically
acceptable salt
thereof, and a PARP inhibitor, or a pharmaceutically acceptable salt thereof.
The present application provides a method of treating a cancer, an autoimmune
disease, an immune deficiency, or a neurodegenerative disease. In some
embodiments, the
method comprises administering to a subject in need thereof a therapeutically
effective

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
amount of a RAD51 inhibitor or a pharmaceutically acceptable salt thereof or a
pharmaceutical composition disclosed herein. In some embodiments, the method
comprises
administering to a subject in need thereof a therapeutically effective amount
of a compound
disclosed herein or a pharmaceutically acceptable salt thereof or a
pharmaceutical
composition disclosed herein.
In one aspect, the present application provides a method of treating a subject
with a
disease which can be ameliorated by inhibition of RAD51 and/or PARP, by
administering to
the subject a therapeutically effective amount of one or more disclosed
compounds, or a
pharmaceutically acceptable salt thereof, or the corresponding pharmaceutical
composition,
in combination with a PARP inhibitor, or a pharmaceutically acceptable salt
thereof, or a
pharmaceutical composition thereof. Diseases which can be ameliorated by
inhibition of
RAD51 and/or PARP include treating cancer, autoimmune disease, immune
deficiency, or
neurodegenerative disease.
In one aspect, described herein is a method of treating cancer, autoimmune
disease,
immune deficiency, or neurodegenerative disease, the method comprising
administering a
therapeutically effective dose of a compound of disclosed herein or a
pharmaceutically
acceptable salt thereof or a pharmaceutical composition disclosed herein in
combination with
a PARP inhibitor or a pharmaceutically acceptable salt thereof or a
pharmaceutical
composition comprising a PARP inhibitor, to a subject in need of treatment for
cancer,
autoimmune disease, immune deficiency, or neurodegenerative disease.
In some embodiments, the present application provides a method of treating a
cancer,
an autoimmune disease, an immune deficiency, or a neurodegenerative disease,
wherein the
method comprises administering to a subject in need thereof a therapeutically
effective
amount of a compound disclosed herein or a pharmaceutically acceptable salt
thereof or a
pharmaceutical composition disclosed herein in combination with a PARP
inhibitor or a
pharmaceutically acceptable salt thereof or a pharmaceutical composition
comprising a PARP
inhibitor.
In some embodiments, the present application provides a method of treating a
cancer,
an autoimmune disease, an immune deficiency, or a neurodegenerative disease,
wherein the
method comprises administering to a subject in need thereof a composition
comprising a
compound disclosed herein, or a pharmaceutically acceptable salt thereof, and
a PARP
inhibitor, or a pharmaceutically acceptable salt thereof.
41

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
In some embodiments, the present application provides a method of treating a
cancer,
an autoimmune disease, an immune deficiency, or a neurodegenerative disease,
wherein the
method comprises administering to a subject in need thereof a combination of a
compound
disclosed herein, or a pharmaceutically acceptable salt thereof, and a PARP
inhibitor, or a
pharmaceutically acceptable salt thereof.
In some embodiments, the present application provides a method of treating a
cancer,
an autoimmune disease, an immune deficiency, or a neurodegenerative disease,
wherein the
method comprises administering to a subject in need thereof a therapeutically
effective
amount of a RAD51 inhibitor or a pharmaceutically acceptable salt thereof or a

pharmaceutical composition disclosed herein in combination with a PARP
inhibitor or a
pharmaceutically acceptable salt thereof or a pharmaceutical composition
comprising a PARP
inhibitor.
In some embodiments, the present application provides a method of treating a
cancer,
an autoimmune disease, an immune deficiency, or a neurodegenerative disease,
wherein the
method comprises administering to a subject in need thereof a composition
comprising a
RAD51 inhibitor, or a pharmaceutically acceptable salt thereof, and a PARP
inhibitor, or a
pharmaceutically acceptable salt thereof.
In some embodiments, the present application provides a method of treating a
cancer,
an autoimmune disease, an immune deficiency, or a neurodegenerative disease,
wherein the
method comprises administering to a subject in need thereof a combination of a
RAD51
inhhibitor, or a pharmaceutically acceptable salt thereof, and a PARP
inhibitor, or a
pharmaceutically acceptable salt thereof.
In some embodiments, the present application provides a method of treating a
cancer,
an autoimmune disease, an immune deficiency, or a neurodegenerative disease,
wherein the
method comprises administering to a subject in need thereof a therapeutically
effective
amount of Compound 67A or a pharmaceutically acceptable salt thereof or a
pharmaceutical
composition disclosed herein in combination with a PARP inhibitor or a
pharmaceutically
acceptable salt thereof or a pharmaceutical composition comprising a PARP
inhibitor.
In some embodiments, the present application provides a method of treating a
cancer,
an autoimmune disease, an immune deficiency, or a neurodegenerative disease,
wherein the
method comprises administering to a subject in need thereof a composition
comprising
Compound 67A, or a pharmaceutically acceptable salt thereof, and a PARP
inhibitor, or a
pharmaceutically acceptable salt thereof.
42

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
In some embodiments, the present application provides a method of treating a
cancer,
an autoimmune disease, an immune deficiency, or a neurodegenerative disease,
wherein the
method comprises administering to a subject in need thereof a combination of
Compound
67A, or a pharmaceutically acceptable salt thereof, and a PARP inhibitor, or a

pharmaceutically acceptable salt thereof.
In some embodiments, the present application provides a method of treating a
cancer,
an autoimmune disease, an immune deficiency, or a neurodegenerative disease,
wherein the
method comprises administering to a subject in need thereof a therapeutically
effective
amount of a compound of the present disclosure or a pharmaceutically
acceptable salt thereof
or a pharmaceutical composition disclosed herein in combination with olaparib
or a
pharmaceutically acceptable salt thereof or a pharmaceutical composition
comprising
olaparib.
In some embodiments, the present application provides a method of treating a
cancer,
an autoimmune disease, an immune deficiency, or a neurodegenerative disease,
wherein the
method comprises administering to a subject in need thereof a composition
comprising a
RAD51 inhibitor, or a pharmaceutically acceptable salt thereof, and olaparib,
or a
pharmaceutically acceptable salt thereof.
In some embodiments, the present application provides a method of treating a
cancer,
an autoimmune disease, an immune deficiency, or a neurodegenerative disease,
wherein the
method comprises administering to a subject in need thereof a combination of a
RAD51
inhhibitor, or a pharmaceutically acceptable salt thereof, and olaparib, or a
pharmaceutically
acceptable salt thereof.
In some embodiments, the present application provides a method of treating a
cancer,
an autoimmune disease, an immune deficiency, or a neurodegenerative disease,
wherein the
method comprises administering to a subject in need thereof a therapeutically
effective
amount of Compound 67A or a pharmaceutically acceptable salt thereof or a
pharmaceutical
composition disclosed herein in combination with olaparib or a
pharmaceutically acceptable
salt thereof or a pharmaceutical composition comprising olaparib.
In some embodiments, the present application provides a method of treating a
cancer,
an autoimmune disease, an immune deficiency, or a neurodegenerative disease,
wherein the
method comprises administering to a subject in need thereof a composition
comprising
Compound 67A, or a pharmaceutically acceptable salt thereof, and olaparib, or
a
pharmaceutically acceptable salt thereof.
43

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
In some embodiments, the present application provides a method of treating a
cancer,
an autoimmune disease, an immune deficiency, or a neurodegenerative disease,
wherein the
method comprises administering to a subject in need thereof a combination of
Compound
67A, or a pharmaceutically acceptable salt thereof, and olaparib, or a
pharmaceutically
acceptable salt thereof.
In some embodiments, the present application provides the use of a compound
disclosed herein, or a pharmaceutically acceptable salt thereof, or a
pharmaceutical
composition disclosed herein in the manufacture of a medicament for the
treatment of a
disease or disorder.
In some embodiments, the present application provides the use of a composition

disclosed herein in the manufacture of a medicament for the treatment of a
disease or
disorder.
In some embodiments, the present application provides the use of a combination

disclosed herein in the manufacture of a medicament for the treatment of a
disease or
disorder.
In some embodiments, the present application provides the use of a compound
disclosed herein, or a pharmaceutically acceptable salt thereof, or a
pharmaceutical
composition disclosed herein in the manufacture of a medicament for the
treatment of a
cancer, an autoimmune disease, an immune deficiency, or a neurodegenerative
disease.
In some embodiments, the present application provides use of a compound
disclosed
herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical
composition
disclosed herein in combination with a PARP inhibitor, or a pharmaceutically
acceptable salt
thereof, or a pharmaceutical composition thereof in the manufacture of a
medicament for the
treatment of a cancer, an autoimmune disease, an immune deficiency, or a
neurodegenerative
disease.
In some embodiments, the present application provides use of a combination of
a
compound disclosed herein, or a pharmaceutically acceptable salt thereof, and
a PARP
inhibitor, or a pharmaceutically acceptable salt thereof in the manufacture of
a medicament
for the treatment of a cancer, an autoimmune disease, an immune deficiency, or
a
neurodegenerative disease.
In some embodiments, the present application provides use of a composition
comprising a compound disclosed herein, or a pharmaceutically acceptable salt
thereof, and a
PARP inhibitor, or a pharmaceutically acceptable salt thereof in the
manufacture of a
44

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
medicament for the treatment of a cancer, an autoimmune disease, an immune
deficiency, or
a neurodegenerative disease.
In some embodiments, the present application provides use of a RAD51
inhibitor, or a
pharmaceutically acceptable salt thereof, or a pharmaceutical composition
disclosed herein in
combination with a PARP inhibitor, or a pharmaceutically acceptable salt
thereof, or a
pharmaceutical composition thereof in the manufacture of a medicament for the
treatment of
a cancer, an autoimmune disease, an immune deficiency, or a neurodegenerative
disease.
In some embodiments, the present application provides use of a combination of
a
RAD51 inhibitor, or a pharmaceutically acceptable salt thereof, and a PARP
inhibitor, or a
pharmaceutically acceptable salt thereof in the manufacture of a medicament
for the
treatment of a cancer, an autoimmune disease, an immune deficiency, or a
neurodegenerative
disease.
In some embodiments, the present application provides use of a composition
comprising a RAD51 inhibitor, or a pharmaceutically acceptable salt thereof,
and a PARP
inhibitor, or a pharmaceutically acceptable salt thereof in the manufacture of
a medicament
for the treatment of a cancer, an autoimmune disease, an immune deficiency, or
a
neurodegenerative disease.
In some embodiments, the present application provides use of Compound 67A, or
a
pharmaceutically acceptable salt thereof, or a pharmaceutical composition
disclosed herein in
combination with a PARP inhibitor, or a pharmaceutically acceptable salt
thereof, or a
pharmaceutical composition thereof in the manufacture of a medicament for the
treatment of
a cancer, an autoimmune disease, an immune deficiency, or a neurodegenerative
disease.
In some embodiments, the present application provides use of a combination of
Compound 67A, or a pharmaceutically acceptable salt thereof, and a PARP
inhibitor, or a
pharmaceutically acceptable salt thereof in the manufacture of a medicament
for the
treatment of a cancer, an autoimmune disease, an immune deficiency, or a
neurodegenerative
disease.
In some embodiments, the present application provides use of a composition
comprising Compound 67A, or a pharmaceutically acceptable salt thereof, and a
PARP
inhibitor, or a pharmaceutically acceptable salt thereof in the manufacture of
a medicament
for the treatment of a cancer, an autoimmune disease, an immune deficiency, or
a
neurodegenerative disease.

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
In some embodiments, the present application provides use of a compound
disclosed
herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical
composition
disclosed herein in combination with olaparib, or a pharmaceutically
acceptable salt thereof,
or a pharmaceutical composition thereof in the manufacture of a medicament for
the
treatment of a cancer, an autoimmune disease, an immune deficiency, or a
neurodegenerative
disease.
In some embodiments, the present application provides use of a combination of
a
compound disclosed herein, or a pharmaceutically acceptable salt thereof, and
olaparib, or a
pharmaceutically acceptable salt thereof in the manufacture of a medicament
for the
treatment of a cancer, an autoimmune disease, an immune deficiency, or a
neurodegenerative
disease.
In some embodiments, the present application provides use of a composition
comprising a compound disclosed herein, or a pharmaceutically acceptable salt
thereof, and
olaparib, or a pharmaceutically acceptable salt thereof in the manufacture of
a medicament
for the treatment of a cancer, an autoimmune disease, an immune deficiency, or
a
neurodegenerative disease.
In some embodiments, the present application provides use of a RAD51
inhibitor, or a
pharmaceutically acceptable salt thereof, or a pharmaceutical composition
disclosed herein in
combination with olaparib, or a pharmaceutically acceptable salt thereof, or a
pharmaceutical
composition thereof in the manufacture of a medicament for the treatment of a
cancer, an
autoimmune disease, an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides use of a combination of
a a
RAD51 inhibitor, or a pharmaceutically acceptable salt thereof, and olaparib,
or a
pharmaceutically acceptable salt thereof in the manufacture of a medicament
for the
treatment of a cancer, an autoimmune disease, an immune deficiency, or a
neurodegenerative
disease.
In some embodiments, the present application provides use of a composition
comprising a a RAD51 inhibitor, or a pharmaceutically acceptable salt thereof,
and olaparib,
or a pharmaceutically acceptable salt thereof in the manufacture of a
medicament for the
treatment of a cancer, an autoimmune disease, an immune deficiency, or a
neurodegenerative
disease.
In some embodiments, the present application provides use of Compound 67A, or
a
pharmaceutically acceptable salt thereof, or a pharmaceutical composition
disclosed herein in
46

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
combination with olaparib, or a pharmaceutically acceptable salt thereof, or a
pharmaceutical
composition thereof in the manufacture of a medicament for the treatment of a
cancer, an
autoimmune disease, an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides use of a combination of
Compound 67A, or a pharmaceutically acceptable salt thereof, and olaparib, or
a
pharmaceutically acceptable salt thereof in the manufacture of a medicament
for the
treatment of a cancer, an autoimmune disease, an immune deficiency, or a
neurodegenerative
disease.
In some embodiments, the present application provides use of a composition
comprising Compound 67A, or a pharmaceutically acceptable salt thereof, and
olaparib, or a
pharmaceutically acceptable salt thereof in the manufacture of a medicament
for the
treatment of a cancer, an autoimmune disease, an immune deficiency, or a
neurodegenerative
disease.
In some embodiments, the present application provides the use of a compound
disclosed herein, or a pharmaceutically acceptable salt thereof, or a
pharmaceutical
composition disclosed herein for the treatment of a disease or disorder.
In some embodiments, the present application provides the use of a composition

disclosed herein for the treatment of a disease or disorder.
In some embodiments, the present application provides the use of a combination

disclosed herein for the treatment of a disease or disorder.
In some embodiments, the present application provides the use of a compound
disclosed herein, or a pharmaceutically acceptable salt thereof, or a
pharmaceutical
composition disclosed herein for the treatment of a cancer, an autoimmune
disease, an
immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides use of a compound
disclosed
herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical
composition
disclosed herein in combination with a PARP inhibitor, or a pharmaceutically
acceptable salt
thereof, or a pharmaceutical composition thereof for the treatment of a
cancer, an
autoimmune disease, an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides use of a combination of
a
compound disclosed herein, or a pharmaceutically acceptable salt thereof, and
a PARP
inhibitor, or a pharmaceutically acceptable salt thereof for the treatment of
a cancer, an
autoimmune disease, an immune deficiency, or a neurodegenerative disease.
47

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
In some embodiments, the present application provides use of a composition
comprising a compound disclosed herein, or a pharmaceutically acceptable salt
thereof, and a
PARP inhibitor, or a pharmaceutically acceptable salt thereof for the
treatment of a cancer, an
autoimmune disease, an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides use of a RAD51
inhibitor, or a
pharmaceutically acceptable salt thereof, or a pharmaceutical composition
disclosed herein in
combination with a PARP inhibitor, or a pharmaceutically acceptable salt
thereof, or a
pharmaceutical composition thereof for the treatment of a cancer, an
autoimmune disease, an
immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides use of a combination of
a
RAD51 inhibitor, or a pharmaceutically acceptable salt thereof, and a PARP
inhibitor, or a
pharmaceutically acceptable salt thereof for the treatment of a cancer, an
autoimmune
disease, an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides use of a composition
comprising a RAD51 inhibitor, or a pharmaceutically acceptable salt thereof,
and a PARP
inhibitor, or a pharmaceutically acceptable salt thereof for the treatment of
a cancer, an
autoimmune disease, an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides use of Compound 67A, or
a
pharmaceutically acceptable salt thereof, or a pharmaceutical composition
disclosed herein in
combination with a PARP inhibitor, or a pharmaceutically acceptable salt
thereof, or a
pharmaceutical composition thereof for the treatment of a cancer, an
autoimmune disease, an
immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides use of a combination of
Compound 67A, or a pharmaceutically acceptable salt thereof, and a PARP
inhibitor, or a
pharmaceutically acceptable salt thereof for the treatment of a cancer, an
autoimmune
disease, an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides use of a composition
comprising Compound 67A, or a pharmaceutically acceptable salt thereof, and a
PARP
inhibitor, or a pharmaceutically acceptable salt thereof for the treatment of
a cancer, an
autoimmune disease, an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides use of a compound
disclosed
herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical
composition
disclosed herein in combination with olaparib, or a pharmaceutically
acceptable salt thereof,
48

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
or a pharmaceutical composition thereof for the treatment of a cancer, an
autoimmune
disease, an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides use of a combination of
a
compound disclosed herein, or a pharmaceutically acceptable salt thereof, and
olaparib, or a
pharmaceutically acceptable salt thereof for the treatment of a cancer, an
autoimmune
disease, an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides use of a composition
comprising a compound disclosed herein, or a pharmaceutically acceptable salt
thereof, and
olaparib, or a pharmaceutically acceptable salt thereof for the treatment of a
cancer, an
autoimmune disease, an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides use of a RAD51
inhibitor, or a
pharmaceutically acceptable salt thereof, or a pharmaceutical composition
disclosed herein in
combination with olaparib, or a pharmaceutically acceptable salt thereof, or a
pharmaceutical
composition thereof for the treatment of a cancer, an autoimmune disease, an
immune
deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides use of a combination of
a a
RAD51 inhibitor, or a pharmaceutically acceptable salt thereof, and olaparib,
or a
pharmaceutically acceptable salt thereof for the treatment of a cancer, an
autoimmune
disease, an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides use of a composition
comprising a a RAD51 inhibitor, or a pharmaceutically acceptable salt thereof,
and olaparib,
or a pharmaceutically acceptable salt thereof for the treatment of a cancer,
an autoimmune
disease, an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides use of Compound 67A, or
a
pharmaceutically acceptable salt thereof, or a pharmaceutical composition
disclosed herein in
combination with olaparib, or a pharmaceutically acceptable salt thereof, or a
pharmaceutical
composition thereof for the treatment of a cancer, an autoimmune disease, an
immune
deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides use of a combination of
Compound 67A, or a pharmaceutically acceptable salt thereof, and olaparib, or
a
pharmaceutically acceptable salt thereof for the treatment of a cancer, an
autoimmune
disease, an immune deficiency, or a neurodegenerative disease.
49

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
In some embodiments, the present application provides use of a composition
comprising Compound 67A, or a pharmaceutically acceptable salt thereof, and
olaparib, or a
pharmaceutically acceptable salt thereof for the treatment of a cancer, an
autoimmune
disease, an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides a compound disclosed
herein,
or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition
disclosed
herein for use in treating a disease or disorder.
In some embodiments, the present application provides a composition disclosed
herein for use in treating a disease or disorder.
In some embodiments, the present application provides a combination disclosed
herein for use in treating a disease or disorder.
In some embodiments, the present application provides a compound disclosed
herein,
or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition
disclosed
herein for use in treating a cancer, an autoimmune disease, an immune
deficiency, or a
neurodegenerative disease.
In some embodiments, the present application provides a compound disclosed
herein,
or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition
disclosed
herein in combination with a PARP inhibitor, or a pharmaceutically acceptable
salt thereof,
or a pharmaceutical composition thereof for use in treating a cancer, an
autoimmune disease,
an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides a combination of a
compound
disclosed herein, or a pharmaceutically acceptable salt thereof and a PARP
inhibitor, or a
pharmaceutically acceptable salt thereof for use in treating a cancer, an
autoimmune disease,
an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides a composition comprising
a
compound disclosed herein, or a pharmaceutically acceptable salt thereof, and
a PARP
inhibitor, or a pharmaceutically acceptable salt thereof for use in treating a
cancer, an
autoimmune disease, an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides a RAD51 inhibitor, or a
pharmaceutically acceptable salt thereof, or a pharmaceutical composition
disclosed herein in
combination with a PARP inhibitor, or a pharmaceutically acceptable salt
thereof, or a
pharmaceutical composition thereof for use in treating a cancer, an autoimmune
disease, an
immune deficiency, or a neurodegenerative disease.

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
In some embodiments, the present application provides a combination of a RAD51

inhibitor, or a pharmaceutically acceptable salt thereof and a PARP inhibitor,
or a
pharmaceutically acceptable salt thereof for use in treating a cancer, an
autoimmune disease,
an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides a composition comprising
a
RAD51 inhibitor, or a pharmaceutically acceptable salt thereof, and a PARP
inhibitor, or a
pharmaceutically acceptable salt thereof for use in treating a cancer, an
autoimmune disease,
an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides Compound 67A, or a
pharmaceutically acceptable salt thereof, or a pharmaceutical composition
disclosed herein in
combination with a PARP inhibitor, or a pharmaceutically acceptable salt
thereof, or a
pharmaceutical composition thereof for use in treating a cancer, an autoimmune
disease, an
immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides a combination of
Compound
67A, or a pharmaceutically acceptable salt thereof and a PARP inhibitor, or a
pharmaceutically acceptable salt thereof for use in treating a cancer, an
autoimmune disease,
an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides a composition comprising

Compound 67A, or a pharmaceutically acceptable salt thereof, and a PARP
inhibitor, or a
pharmaceutically acceptable salt thereof for use in treating a cancer, an
autoimmune disease,
an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides a compound disclosed
herein,
or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition
disclosed
herein in combination with olaparib, or a pharmaceutically acceptable salt
thereof, or a
pharmaceutical composition thereof for use in treating a cancer, an autoimmune
disease, an
immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides a combination of a
compound
disclosed herein, or a pharmaceutically acceptable salt thereof and olaparib,
or a
pharmaceutically acceptable salt thereof for use in treating a cancer, an
autoimmune disease,
an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides a composition comprising
a
compound disclosed herein, or a pharmaceutically acceptable salt thereof, and
olaparib, or a
51

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
pharmaceutically acceptable salt thereof for use in treating a cancer, an
autoimmune disease,
an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides a RAD51 inhibitor, or a
pharmaceutically acceptable salt thereof, or a pharmaceutical composition
disclosed herein in
combination with olaparib, or a pharmaceutically acceptable salt thereof, or a
pharmaceutical
composition thereof for use in treating a cancer, an autoimmune disease, an
immune
deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides a combination of a RAD51

inhibitor, or a pharmaceutically acceptable salt thereof and olaparib, or a
pharmaceutically
acceptable salt thereof for use in treating a cancer, an autoimmune disease,
an immune
deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides a composition comprising
a
RAD51 inhibitor, or a pharmaceutically acceptable salt thereof, and olaparib,
or a
pharmaceutically acceptable salt thereof for use in treating a cancer, an
autoimmune disease,
an immune deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides Compound 67A, or a
pharmaceutically acceptable salt thereof, or a pharmaceutical composition
disclosed herein in
combination with olaparib, or a pharmaceutically acceptable salt thereof, or a
pharmaceutical
composition thereof for use in treating a cancer, an autoimmune disease, an
immune
deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides a combination of
Compound
67A, or a pharmaceutically acceptable salt thereof and olaparib, or a
pharmaceutically
acceptable salt thereof for use in treating a cancer, an autoimmune disease,
an immune
deficiency, or a neurodegenerative disease.
In some embodiments, the present application provides a composition comprising

Compound 67A, or a pharmaceutically acceptable salt thereof, and olaparib, or
a
pharmaceutically acceptable salt thereof for use in treating a cancer, an
autoimmune disease,
an immune deficiency, or a neurodegenerative disease.
In some embodiments, the disease or disorder is a cancer.
In some embodiments, the disease or disorder is an autoimmune disease.
In some embodiments, the disease or disorder is an immune deficiency.
In some embodiments, the disease or disorder ia a neurodegenerative disease.
In some embodiments, a compound disclosed herein is a RAD51 inhibitor.
52

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
In some embodiments, the RAD51 inibitor is compound 67A.
In some embodiments, the PARP inhibitor is olaparib.
In some embodiments, the disease or disorder is a disease or disorder in which

RAD51 plays a role in the initiation or development of the disease or
disorder. In some
embodiments, the disease or disorder is a disease or disorder in which PARP
plays a role in
the initiation or development of the disease or disorder. In some embodiments,
the disease or
disorder is a disease or disorder in which RAD51 or PARP plays a role in the
initiation or
development of the disease or disorder. In some embodiments, the disease or
disorder is a
disease or disorder in which RAD51 and PARP play a role in the initiation or
development of
the disease or disorder.
In some embodiments, the disease or disorder is cancer in which RAD51 plays a
role
in the initiation or development of the cancer. In some embodiments, the
disease or disorder
is cancer in which PARP plays a role in the initiation or development of the
cancer. In some
embodiments, the disease or disorder is cancer in which RAD51 or PARP plays a
role in the
initiation or development of the cancer. In some embodiments, the disease or
disorder is
cancer in which RAD51 and PARP play a role in the initiation or development of
the disease
or disorder.
In some embodiments, the disease or disorder is associated with (e.g.,
possesses) one
or more mutations in or dysregulation of, or is deficient in, one or more
genes selected from
BRCA1, BRCA2, ATR, ATM, CHK1, CHK2, RAD51, RPA, XRCC3, FANCA, FANCC,
FANCD2, FANCF, FANCG, and FANCM. In some embodiments, the disease or disorder
is
cancer associated with (e.g., possesses) one or more mutations in or
dysregulation of, or is
deficient in, one or more genes selected from BRCA1, BRCA2, ATR, ATM, CHK1,
CHK2,
RAD51, RPA, XRCC3, FANCA, FANCC, FANCD2, FANCF, FANCG, and FANCM.
In some embodiments, the cancer described herein is selected from
hematological
cancer, breast cancer, ovarian cancer, pancreatic cancer, prostate cancer,
fallopian tube
cancer, peritoneal cancer, and lung cancer, each as described herein. In some
embodiments,
the cancer described herein is a recurrent cancer (e.g, recurrent breast
cancer, or recurrent
ovarian cancer). In some embodiments, the cancer described herein is a
metastatic cancer. In
some embodiments, the cancer described herein is associated with (e.g.,
possesses) one or
more mutations in or dysregulation of, or is deficient in, one or more genes
selected from
BRCA1, BRCA2, ATR, ATM, CHK1, CHK2, RAD51, RPA, XRCC3, FANCA, FANCC,
FANCD2, FANCF, FANCG, and FANCM. In some embodiments, the cancer described
53

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
herein is associated with (e.g., possesses) one or more mutations in or
dysregulation of, or is
deficient in BRCA1 and/or BRCA2.
In some embodiments, the subject can be a subject determined to have an
increased
level of DNA damage occurring in one or more cell types relative to a
reference level. As
used herein, "DNA damage" refers to breaks, nicks, and mutations of the DNA
present in a
cell. In some embodiments, the DNA damage can comprise one or more of single-
strand
breaks (e.g., "nicks"), double strand breaks (DSBs), and mutations. In some
embodiments, the
DNA damage can be one or more DSBs. As used herein, "mutation" refers to a
change or
difference in the genetic material of a cell as compared to a reference
wildtype cell, e.g. a
deletion, an insertion, a SNP, a gene rearrangement, and/or the introduction
of an exogenous
gene or sequence.
In some embodiments, the subject can be determined to have an increased level
of
DNA damage if the subject is determined to have an increased level and/or
activity of a DNA
damage process or DNA editing enzyme. As used herein, "DNA damage process"
refers to
any activity or process in a cell which causes one or more types of DNA damage
to occur.
In some embodiments, an increased level of DNA damage can be an increased
level
of mutations, e.g., by determining the overall mutation status in all or a
portion of the genome
of a cell. An overall mutation status at least 2% greater, e.g. 2% greater or
more, 3% greater
or more, 5% greater or more, 10% greater or more, or 20% greater or more than
the overall
mutation status in a reference cell can be indicative of an increased,
elevated, and/or
significant level of a DNA editing enzyme activity. In some embodiments, the
level of hyper
mutations can be determined. In some embodiments, the overall mutation status
in the whole
genome or a portion thereof can be determined using FISH, whole genome
sequencing, high
throughput sequencing, exome sequencing, hybridization, and/or PCR. In some
embodiments
the activity of a DNA editing enzyme can be measured by determining the level
of
hypermutations in the specific target genes including, but not limited to IGH,
BCL6, MYC,
BCL1 1A, CD93, PIM1 and/or PAX5. In some embodiments the DNA editing enzyme is
AID.
In some embodiments, a level of mutation in specific target genes including
IGH, BCL6,
MYC, BCL1 1A, CD93, PIM1 and/or PAX5 which is at least 2% greater, e.g. 2%
greater or
more, 3% greater or more, 5% greater or more, 10% greater or more, or 20%
greater or more
than the level of mutation in IGH, BCL6, MYC, BCL1 1A, CD93, PIM1 and/or PAX5
in a
reference cell can be indicative of an increased, elevated, and/or significant
level of AID
activity.
54

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
In some embodiments, an increased level of DNA damage can be an increased
level
of double strand breaks (DSBs). The level of DSBs can be determined, by way of
non-
limiting example, by karyotyping, by y-H2AX foci formation, and/or by using
FISH analysis
to detect DNA double strand breaks, e.g. DNA breakage detection fish (DBD-
FISH) (Volpi
and Bridger, BioTechniques, Vol. 45, No. 4, October 2008, pp. 385-409).
In some embodiments, an increased level of DNA damage can be an increased
level
of single strand breaks. The level of single-strand breaks in DNA can be
determined, by way
of non-limiting example, by COMET assays, FISH, or the use of single-strand
break-specific
probes. Detection of DNA breaks, both single and double -stranded is known in
the art and
described further, at, e.g., Kumari et al. EXCLI Journal 2009 7:44-62 and
Motalleb et al.
Research Journal of Applied Sciences, Engineering and Technology. 2012 4: 1888-
1894;
each of which is incorporated by reference herein in its entirety.
In some embodiments, an increased level of activity of a DNA damage process
can
comprise an increased level and/or activity of a DNA editing enzyme. In some
embodiments,
the technology described herein is directed to treating cells having an active
DNA editing
enzyme with a compound of the present application in combination with a PARP
inhibitor. In
some embodiments, the technology described herein is directed to treating
cells having an
increased level and/or activity of a DNA editing enzyme with a compound of the
present
application in combination with a PARP inhibitor. As used herein, "DNA editing
enzyme"
refers to an enzyme which normally catalyzes the mutation, exchange or
excision of DNA
segments, particularly enzymes which can generate or promote the generation of
point
mutations, DNA single strand breaks, DNA double-strand breaks or protein-DNA
adducts. A
DNA editing enzyme, as referred to herein, is not necessarily site-specific in
its action.
Similarly, it is not necessarily cell specific. In some embodiments, the cell
is a B cell
expressing a detectable amount of such an enzyme.
Non-limiting examples of DNA editing enzymes include, but are not limited to
Recombination Activating Gene 1 (RAG1; NCBI Gene ID: 5896), Recombination
Activating
Gene 1 (RAG2; NCBI Gene ID: 5897), Sporulation-specific protein 11 (SPO1 1;
NCBI Gene
ID: 23626), APOBEC family members a Type 1 Topoisomerase; a Type 2
Topoisomerase;
and/or AID. In some embodiments, the DNA editing enzyme can be AID.
In some embodiments, the DNA editing enzyme can be a member of the APOBEC
(apolipoprotein B mRNA editing enzyme, catalytic polypeptide -like) family. As
used herein
"APOBEC family" refers to a family of cytidine deaminase enzymes having an N-
terminal

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
zinc-dependent cytidine deaminase catalytic domain comprising and a C-terminal

pseudocatalytic domain. Non-limiting examples of APOBEC family members include
AID,
APOBEC 1 (e.g., NCBI Gene ID: 339), APOBEC2 (e.g., NCBI Gene ID: 10930),
APOBEC3A (e.g., NCBI Gene ID: 200315), APOBEC3C (e.g., NCBI Gene ID: 27350),
APOBEC3E (e.g., NCBI Gene ID: 140564), APOBEC3F (e.g., NCBI Gene ID:200316),
APOBEC3G (e.g., NCBI Gene ID: 60489), APOBEC3H (e.g., NCBI Gene ID: 164668),
and
APOBEC4(e.g., NCBI Gene ID: 403314).
In some embodiments, the DNA editing enzyme can be a Type 1 topoisomerase. In
some embodiments, the DNA editing enzyme can be a Type 2 topoisomerase.
Topoisomerases generate breaks in DNA to help uncoil or relax the strand. Type
II
topoisomerases hydrolyze ATP to generate DSB cuts, while Type I topoisomerases
generate
single-stranded breaks. Non-limiting examples of Type II topoisomerases can
include
topoisomerase II (e.g., NCBI Gene ID: 7153 and 7155). Non-limiting examples of
Type I
topoisomerases can include topoisomerase I (e.g., NCBI Gene ID: 7150).
Embodiments of the technology described herein are based on the discovery that
the
compounds described herein can inhibit DNA repair mechanisms, e.g., homologous
repair,
and act synergistically with a PARP inhibitor. Activation-induced cytidine
deaminase (AID,
or AICDA, also known as ARP2, CDA2 or HIGM2), a DNA-editing enzyme that is a
member of the apolipoprotein B mRNA editing enzymes, catalytic polypeptide -
like
(APOBEC), will cause widespread genomic breaks and cell death in cells with
diminished
homologous recombination ability (e.g. cells with diminished DNA double strand
break
repair abilities). Accordingly, provided herein is a method of causing cell
death comprising
detecting increased expression of a DNA-editing enzyme (e.g. AID) in a cell
and thereafter
contacting the cell with a compound of the present application in combination
with a PARP
inhibitor; thereby resulting in cell death. Accordingly, provided herein is a
method of causing
cell death comprising increasing expression of a DNA-editing enzyme (e.g. AID)
in a cell
and thereafter contacting the cell with a compound of the present application
in combination
with a PARP inhibitor; thereby resulting in cell death. Accordingly, provided
herein is a
method of causing cell death comprising administering to a cell a
therapeutically effective
amount of a DNA editing enzyme (e.g. AID) and thereafter contacting the cell
with a
compound of the present application in combination with a PARP inhibitor;
thereby resulting
in cell death.
56

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
AID, encoded by the AICDA gene (NCBI Gene ID: 57379), is required for proper B-

cell function and is most prominently expressed in centroblast B-cells. The
protein is
involved in somatic hypermutation, gene conversion, and class-switch
recombination of
immunoglobulin genes. AID is normally expressed almost exclusively in antigen-
activated
germinal center B-cells, where it initiates immunoglobulin isotype class
switching (Manis et
al. 2002, Trends Immunol, 23, 31-39; Chaudhuri and Alt, Nat Rev Immunol, 2004,
4, 541-
552; Longerich et al., Curr Opin Immunol, 2006, 18, 164-174; Chaudhuri et al.,
Adv
Immunol 2007, 94, 157-214). AID is required for somatic hypermutation and
immunoglobulin class switching in activated B cells. AID expression is
regulated by CD40
ligand, B-cell receptor, IL4R, or Toll-like receptor stimulation (Crouch et
al., J Exp Med
2007 204: 1145-1156; Muramatsu et al., J Biol Chem 1999 274: 18470-6). After
activation,
AID is transiently upregulated, induces point mutations or DNA double strand
breaks in a
sequence nonspecific manner within immunoglobulin genes, and is then
downregulated
(Longerich et al., Curr Opin Immunol, 2006, 18, 164-176; Chaudhuri et al., Adv
Immunol
2007, 94, 157-214). Overall, AID is active in only a tiny population of normal
cells (antigen-
activated B-cells) at any given time. The genomic rearrangements and mutations
controlled
by AID lead to the development of antigen-recognition diversity, receptor
editing and
lymphoid effector function required for functional adaptive immunity (Mills,
et al. Immunol
Rev 2003 194:77-95). Recently it has been reported that AID has off-target
point mutation
activities (Liu, M. et al., Nature 2008, 451, 841-845; Liu and Schatz, Trends
Immunol. 2009,
30, 173-181; Perez-Duran et al., Carcinogenesis. 2007, 28(12):2427-33).
Robbiani et al. has
reported off-target activities of AID in B- cells, especially c-myc/IgH
translocations
(Robbiani et al., Mol Cell 2009, 36(4):631-41). AID expression accelerates the
rate of tumor
development in Bc16 transgenic mice (Pasqualucci et al., 2008, Nat. Genet. 40,
108-112).
However, deregulated AID does not necessarily cause malignancy or
translocation-associated
cancer on its own in B cells (Muto et al., 2006, Proc. Natl. Acad. Sci. USA
103, 2752-2757;
Okazaki et al., 2003, J. Exp. Med. 197, 1173-1181; Shen et al., 2008, Mol.
Immunol. 45,
1883-1892). In addition, despite its obligate role in c-myc/IgH translocation,
AID is not
required for the development of plasmacytosis or plasmacytoma in IL-6
transgenic or
pristane-treated mice, respectively (Kovalchuk et al., 2007, J. Exp. Med. 204,
2989-3001;
Ramiro et al., 2004, J. Exp. Med. 200, 1103-1110). However, most human B cell
lymphoma-
associated translocations do not involve c-myc, and many do not involve Ig
genes (Kuppers,
2005, Oncogene 20, 5580-5594).
57

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
Overexpression of AID has been reported in chronic lymphocytic leukemia (CLL)
(Hancer et al. Leuk Lymphoma. 2011 Jan;52(1):79-84; Heintel et al., Leukemia.
2004 Apr;
18(4):756-62). Further, AID expression has been shown to be correlated with
blast crisis B
lineage leukemia and therapy resistance in myeloid leukemia and to be
associated with
generally poor prognosis in chronic B lymphocytic leukemia (Mao et al., Br J
Dermatol 2001,
145: 117-122; Chaudhuri et al., Nature 2004, 430:992-8). Further expression of
AID in tumor
cells from a variety of cancers has been reported including but not limited to
lung, breast,
gastric, colon, intestinal, liver cancer and choriangiocarcinoma (Greeve et
al., Blood 2003,
1010, 3574-3580; Feldhahn et al., J Exp Med 2007, 204, 1157-1166; Kotani et
al., PNAS
USA 2007, 104, 1616-1620; Engels et al., 2008, Appl Immunohistochem Mol
Morphol 16,
521-529; Klemm et al., 2009, Cancer Cell 6, 232-245; Palacios et al., 2010,
Blood 115(22),
4488-4496; Leuenberger et al., 2009, Mod Pathol 32, 177-186; Gruber et al.,
2010, Cancer
Res 70, 7411-7420; inflammatory cancer (Marusawa 2008, Int J Biochem Cell
Bio1.40, 399-
402); follicular lymphoma (Hardianti et al., 2004, Leukemia 18, 826-831;
Shikata et al.,
2012, Cancer Sci. 103(3):415-21); thyroid cancer (Qiu et al. 2012, Mod Pathol
25(0,36-45);
breast cancer (Borchert et al. 2011, BMC Cancer 11:347); Marusawa, et al.,
2011, Adv
Immunol 111: 109-41; Zhang et al. 2012, Hum Pathol 43(3):423-34; Komori et
al., 2008,
Hepatology 47(3):888-896; Hockley 2010, Leukemia 24(5): 1084-6; adult T-cell
leukemia
(Nakamura et al., 2011, Br J Dermatol. 165(2):437-9). All of the references in
the foregoing
paragraph are incorporated by reference herein in their entireties.
Elevated levels of AID have been reported in arthritis (Xu et al. Scand. J.
Immunol.
2009, 296, 2033-6) and in the MRL/Fas(lprilpr) mouse lupus model (White et al.
2011,
Autoimmunity 44(8), 585-98). All of the references in the foregoing paragraph
are
incorporated by reference herein in their entireties.
When DSB repair is inhibited, the extent of the DSBs generated by AID is much
higher than previously suspected and the extent of genomic damage is so severe
as to result in
cell death. Accordingly, in one embodiment of the technology described herein,
there is
provided a method of treatment comprising; (a) selecting a subject having
cells that express
elevated levels of activation-induced cytidine deaminase (AID); and (b)
administering a
therapeutically effective amount of an inhibitor of double strand break repair
(e.g. a
compound of the present application) to the subject; wherein an elevated level
of AID is a
level of AID that is higher than the level of AID in cells of the same type
from a healthy
individual. In some embodiments, the cells expressing elevated levels of AID
are B cells. In
58

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
some embodiments, the B cell expressing elevated levels of AID is a cancerous
B cells or a B
cell associated with autoimmune disease. In some embodiments, the subject can
be a human
subject.
Methods provided herein treat cancers and/or autoimmune disorders by
inhibiting
DNA double strand break repair. This inhibition proves lethal to cells
expressing AID, as
AID generates widespread genomic breaks, and the treatment with a double
strand break
repair inhibitor prevents the repair of these lesions which are being
generated by the cell
itself. This results in cell death in the subject which is specific to the
cells expressing AID,
e.g. cancerous B cells and/or autoimmune cells. Accordingly, as described
herein, in one
embodiment there is a provided a treatment paradigm that selectively induces
self-destruction
of certain diseased cells, while reducing the unintended side effects in
healthy tissues.
In some embodiments, an increased level and/or activity of a DNA editing
enzyme
can be an increased level of DNA editing enzyme mRNA. mRNA levels can be
assessed
using, e.g., biochemical and molecular biology techniques such as Northern
blotting or other
hybridization assays, nuclease protection assay, reverse transcription
(quantitative RT-PCR)
techniques, RNA-Seq, high throughput sequencing and the like. Such assays are
well known
to those in the art. In one embodiment, nuclear "run-on" (or "run-off)
transcription assays are
used (see e.g. Methods in Molecular Biology, Volume: 49, Sep-27-1995, Page
Range: 229-
238). Arrays can also be used; arrays, and methods of analyzing mRNA using
such arrays
have been described previously, e.g. in EP0834575, EP0834576, W096/31622, U.S.
Pat. No.
5,837,832 or W098/30883. W097/10365 provides methods for monitoring of
expression
levels of a multiplicity of genes using high density oligonucleotide arrays.
In some embodiments, a subject can be determined to have an increased level of
DNA
damage occurring in one or more cell types relative to a reference level if
the subject has
been exposed to an agent that is known to cause such DNA damage. Non-limiting
examples
of such agents can include a viral infection with a DNA integrating virus
(e.g. adeno-
associated virus, retrovirus, human T-lymphotropic virus, HIV-1, oncovirus,
hepatitis virus,
hepatitis B virus); DNA damaging chemicals (e.g. acetaldehyde, polycyclic
aromatic
hydrocarbons, benzenes, nitrosamines, tobacco smoke, aflatoxin, and the like);
DNA
damaging chemotherapeutic agents (e.g. bleomycin, mitomycin, nitrogen mustards
(e.g.
mechlorethamine, cyclophosphamide, melphalan, chlorambucil, ifosfamide and
busulfan),
nitrosoureas (e.g., N-Nitroso-N-methylurea (MNU), carmustine (BCNU), lomustine
(CCNU)
and semustine (MeCCNU), fotemustine and streptozotocin), tetrazines (e.g.,
dacarbazine,
59

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
mitozolomide and temozolomide),aziridines (e.g., thiotepa, mytomycin and
diaziquone
(AZQ)), cisplatins (e.g., cisplatin, carboplatin and oxaliplatin) procarbazine
and
hexamethylmelamine); and ionizing or ultraviolet radiation. Exposure to such
agents can be
the result of an accident, infection and/or environmental exposure or the
result of a
therapeutic administration of such agents.
In some embodiments, the increased level of DNA damage can be occurring in a
cell
type affected by the cancer, autoimmune disease, and/or neurodegenerative
disease. In some
embodiments, the subject is determined to have an increased level of DNA
damage occurring
in a cell selected from the group consisting of: a cancer cell; an immune
system cell; or a
nervous system cell.
In some embodiments, the DNA editing enzyme can be AID. In some embodiments,
the level of AID can be the level of AID in a blood cell. In some embodiments,
the level of
AID can be the level of AID in a B cell.
In some embodiments, an increased level of AID can be a detectable level of
AID,
e.g., as described below herein.
In some embodiments, the subject can be a human subject.
Methods provided herein treat cancers and/or autoimmune disorders by
inhibiting
DNA double strand break repair. This inhibition proves lethal to cells
expressing AID, as
AID generates widespread genomic breaks, and the treatment with a double
strand break
repair inhibitor prevents the repair of these lesions which are being
generated by the cell
itself. This results in cell death in the subject which is specific to the
cells expressing AID,
e.g. cancerous B cells and/or autoimmune cells. Accordingly, as described
herein, in one
embodiment there is a provided a treatment paradigm that selectively induces
self-destruction
of certain diseased cells, while reducing the unintended side effects in
healthy tissues.
Methods of defecting cancers in patients with increased levels of DNA damage
or
increased levels of DNA editing enzymes are disclosed in W02016/094897,
incorporated
herein by reference.
In some embodiments, the cancer to be treated is a type with high expression
of a
DNA editing enzyme. In some embodiments, the cancer to be treated is a B-cell
neoplasm.
Another embodiment is a method of treating a cancer by administering to the
subject a
therapeutically effective amount of one or more disclosed compounds, or a
pharmaceutically
acceptable salt thereof, or the corresponding pharmaceutical composition in
combination with
a PARP inhibitor or a pharmaceutically acceptable salt thereof or a
pharmaceutical

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
composition comprising a PARP inhibitor. In one aspect, the cancer is selected
from the
group consisting of lymphoma, leukemia, and a plasma cell neoplasm. In another
aspect, the
cancer selected from the group consisting of carcinoma and sarcoma.
In some embodiments, the cancer to be treated is a lymphoma. Lymphomas which
can
be treated by the disclosed methods include Non-Hodgkin's lymphoma; Burkitt's
lymphoma;
small lymphocytic lymphoma; lymphoplasmacytic lymphoma; MALT lymphoma;
follicular
lymphoma; diffuse large B-cell lymphoma; mantle cell lymphoma; and T-cell
lymphoma.
Lymphoma is a malignancy in the lymphatic cells of the immune system (e.g. B
cells,
T cells, or natural killer (NK) cells). Lymphomas often originate in the lymph
nodes and
present as solid tumors. They can metastasize to other organs such as the
brain, bone, or skin.
Extranodal sites are often located in the abdomen. Lymphomas are closely
related to the
lymphoid leukemia and in some cases a particular form of cancer is categorized
as both a
lymphoma and a leukemia.
Leukemias which can be treated by the disclosed methods include acute
lymphoblastic leukemia (ALL); Burkitt's leukemia; B-cell leukemia; B-cell
acute
lymphoblastic leukemia; chronic lymphocytic leukemia (CLL); acute myelogenous
leukemia
(AML); chronic myelogenous leukemia (CML); and T-cell acute lymphoblastic
leukemia (T-
ALL).
In some embodiments the cancer to be treated is B-cell neoplasms, B-cell
leukemia,
B-cell acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic
myelogenous
leukemia, Burkitt's leukemia, acute myelogenous leukemia and/or T-ALL. The
maturation of
B cells most typically ceases or substantially decreases when the foreign
antigen has been
neutralized. Occasionally, however, proliferation of a particular B cell will
continue
unabated; such proliferation can result in a cancer referred to as "B-cell
lymphoma" or a "B-
cell leukemia." In some embodiments the cancer to be treated is chronic
lymphocytic
leukemia (CLL) or chronic myelogenous leukemia (CML).
In some embodiments the cancer to be treated is a plasma cell neoplasm.
Examples
for plasma cell neoplasms include multiple myeloma; plasma cell myeloma;
plasma cell
leukemia and plasmacytoma.
Carcinomas which can be treated by the disclosed methods include colon cancer;
liver
cancer; gastric cancer; intestinal cancer; esophageal cancer; breast cancer;
ovarian cancer;
head and neck cancer; lung cancer; and thyroid cancer.
61

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
Sarcomas which can be treated by the disclosed methods include soft tissue
sarcoma
and bone sarcoma.
Any cancer characterized by high levels of DNA damage and/or DNA editing
enzyme
expression can be treated with a combination as described herein. For example,
sarcomas,
epithelial cell cancer (carcinomas), colon cancer, gastric cancer, intestinal
cancer, liver
cancer, hepatocellular cancer, breast cancer, thyroid cancer, esophageal
cancer, lung cancer,
brain cancer, head and neck cancer, melanoma, renal cancer, prostate cancer,
hemangioma,
rhabdomyosarcoma, chondrosarcoma, osteosarcoma, fibrosarcoma and
cholangiocarcinoma
may be characterized by high levels of a DNA editing enzyme expression, e.g.
AID. In some
embodiments the cancer to be treated is colon cancer, liver cancer, gastric
cancer, intestinal
cancer, breast cancer, lung cancer, thyroid cancer and/or cholangiocarcinoma.
Specific cancers that can be treated by the disclosed methods include cancer
of the
bladder, blood, bone, bone marrow, brain, breast, colon, esophagus,
gastrointestine, gum,
head, kidney, liver, lung, nasopharynx, neck, ovary, prostate, skin, stomach,
testis, tongue, or
uterus. In addition, the cancer may specifically be of the following
histological type, though it
is not limited to these: neoplasm, malignant; carcinoma; carcinoma,
undifferentiated; giant
and spindle cell carcinoma; sarcomas; small cell carcinoma; papillary
carcinoma; squamous
cell carcinoma; lymphoepithelial carcinoma; basal cell carcinoma; pilomatrix
carcinoma;
transitional cell carcinoma; papillary transitional cell carcinoma;
adenocarcinoma;
gastrinoma, malignant; cholangiocarcinoma; hepatocellular carcinoma; combined
hepatocellular carcinoma and cholangiocarcinoma; trabecular adenocarcinoma;
adenoid
cystic carcinoma; adenocarcinoma in adenomatous polyp; adenocarcinoma,
familial
polyposis coli; solid carcinoma; carcinoid tumor, malignant; branchiolo-
alveolar
adenocarcinoma; papillary adenocarcinoma; chromophobe carcinoma; acidophil
carcinoma;
oxyphilic adenocarcinoma; basophil carcinoma; clear cell adenocarcinoma;
granular cell
carcinoma; follicular adenocarcinoma; papillary and follicular adenocarcinoma;

nonencapsulating sclerosing carcinoma; adrenal cortical carcinoma; endometroid
carcinoma;
skin appendage carcinoma; apocrine adenocarcinoma; sebaceous adenocarcinoma;
ceruminous adenocarcinoma; mucoepidermoid carcinoma; cystadenocarcinoma;
papillary
cystadenocarcinoma; papillary serous cystadenocarcinoma; mucinous
cystadenocarcinoma;
mucinous adenocarcinoma; signet ring cell carcinoma; infiltrating duct
carcinoma; medullary
carcinoma; lobular carcinoma; inflammatory carcinoma; paget's disease,
mammary; acinar
cell carcinoma; adenosquamous carcinoma; adenocarcinoma w/squamous metaplasia;
62

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
thymoma, malignant; ovarian stromal tumor, malignant; thecoma, malignant;
granulosa cell
tumor, malignant; androblastoma, malignant; sertoli cell carcinoma; leydig
cell tumor,
malignant; lipid cell tumor, malignant; paraganglioma, malignant; extra-
mammary
paraganglioma, malignant; pheochromocytoma; glomangiosarcoma; malignant
melanoma;
amelanotic melanoma; superficial spreading melanoma; malig melanoma in giant
pigmented
nevus; epithelioid cell melanoma; blue nevus, malignant; sarcoma;
fibrosarcoma; fibrous
histiocytoma, malignant; myxosarcoma; liposarcoma; leiomyosarcoma;
rhabdomyosarcoma;
embryonal rhabdomyosarcoma; alveolar rhabdomyosarcoma; stromal sarcoma; mixed
tumor,
malignant; mullerian mixed tumor; nephroblastoma; hepatoblastoma;
carcinosarcoma;
mesenchymoma, malignant; brenner tumor, malignant; phyllodes tumor, malignant;
synovial
sarcoma; mesothelioma, malignant; dysgerminoma; embryonal carcinoma; teratoma,

malignant; struma ovarii, malignant; choriocarcinoma; mesonephroma, malignant;

hemangiosarcoma; hemangioendothelioma, malignant; Kaposi's sarcoma;
hemangiopericytoma, malignant; lymphangiosarcoma; osteosarcoma; juxtacortical
osteosarcoma; chondrosarcoma; chondroblastoma, malignant; mesenchymal
chondrosarcoma; giant cell tumor of bone; Ewing's sarcoma; odontogenic tumor,
malignant;
ameloblastic odontosarcoma; ameloblastoma, malignant; ameloblastic
fibrosarcoma;
pinealoma, malignant; chordoma; glioma, malignant; ependymoma; astrocytoma;
protoplasmic astrocytoma; fibrillary astrocytoma; astroblastoma; glioblastoma;

oligodendroglioma; oligodendroblastoma; primitive neuroectodermal; cerebellar
sarcoma;
ganglioneuroblastoma; neuroblastoma; retinoblastoma; olfactory neurogenic
tumor;
meningioma, malignant; neurofibrosarcoma; neurilemmoma, malignant; granular
cell tumor,
malignant; malignant lymphoma; Hodgkin's disease; hodgkin's; paragranuloma;
malignant
lymphoma, small lymphocytic; malignant lymphoma, large cell, diffuse;
malignant
lymphoma, follicular; mycosis fungoides; other specified non-Hodgkin's
lymphomas;
malignant histiocytosis; multiple myeloma; mast cell sarcoma;
immunoproliferative small
intestinal disease; leukemia; lymphoid leukemia; plasma cell leukemia;
erythroleukemia;
lymphosarcoma cell leukemia; myeloid leukemia; basophilic leukemia;
eosinophilic
leukemia; monocytic leukemia; mast cell leukemia; megakaryoblastic leukemia;
myeloid
sarcoma; and hairy cell leukemia.
In another embodiment for the disclosed method, the cancer is characterized by

mutations in the mutS homologues (e.g., MSH2, MSH3, and MSH6), mutL homologues
(e.g.
MLH1), or mismatch repair endonuclease PMS2. Mutations are changes in the
genetic code.
63

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
They include point mutations and frameshift mutations. In a point mutation,
one nucleotide is
swapped out for another. Therefore, the mutation occurs at a single point or
location within
the DNA strand. Frameshift mutations are due to either insertions or deletions
of nucleotides.
This causes the entire DNA strand to elongate or to shrink in size. Thus,
frameshift mutations
may alter all of the codons that occur after the deletion or insertion. The
mutations referred to
herein include, but are not limited to, insertions, deletions, duplications,
inversions, or other
recognized point mutations. It has now been found that RAD51 inhibitors are
particularly
effective in treating cancers with mutations in MSH (e.g. MSH6), MLH, or PMS2.
MutS Homolog 2 (MSH2) is a protein that in humans is encoded by the MSH2 gene,

which is located on chromosome 2. MSH2 is a tumor suppressor gene and more
specifically a
caretaker gene that codes for a DNA mismatch repair (MMR) protein, MSH2, which
forms a
heterodimer with MSH6 to make the human MutSa mismatch repair complex. It also

dimerizes with MSH3 to form the MutSP DNA repair complex. MSH2 is involved in
many
different forms of DNA repair, including transcription-coupled repair,
homologous
recombination, and base excision repair. Examples of the mutations in MSH2
include, but are
not limited to, g.47630253 47630254de1, g.47702411 47702421del,
g.47709913 47709915inv, g.47635629 47635634de1, g.47637227 47637236dup,
g.47639550 47639561del, g.(? 47630206) (47710367 ?)del,
g.(? 47630206) (47643569 47656880)del, g.47630263 47643568de1,
g.(? 47630206) (47657081 47672686)del, g.47630263 47657080de1,
g.(? 47630206) (47672797 47690169)del, g.47630263 47672796de1,
g.(? 47630206) (47672797 47690169)del, g.(? 47630206) (47693948 47698103)del,
g.47630263 47693947de1, g.(? 47630206) (47698202 47702163)del,
g.(? 47630206) (47630542 47635539)del, g.(? 47630206) (47708011 47709917)del,
g.(? 47630206) (47635695 47637232)del, g.(? 47630206) (47635695 47637232)del,
g.(? 47630206) (47637512 47639552)del, g.(? 47630206) (47639700 47641407)del,
g.(? 47630206) (47641558 47643434)del, g.47618487 47650860delins(155),
g.47628578 47638433del, g.47595033 47662777de1, g.47583175 47667707del,
g.47625602 47636880de1, g.47554933 47699909de1, g.47629508 47649552de1,
g.47629375 47651274de1, g.(? 47630206) (47630542 47635539)del,
g.(? 47630206) (47635695 47637232)del, g.47643509 47643510de1,
g.47643529 47643530dup, g.47656746 47657199dup, g.47656661 47663325del,
g.(47643569 47656880) (47710367 ?)del, g.(47643569 47656880) (47710367 ?)del,
64

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
g.47656881 47657080del, g.(47643569 47656880) (47657081 47672686)del,
g.(47643569 47656880) (47657081 47672686)del,
g.(47643569 47656880) (47657081 47672686)del,
g.(47643569 47656880) (47657081 47672686)dup,
g.(47643569 47656880) (47657081 47672686)dup,
g.(47643569 47656880) (47672797 47690169)del,
g.(47643569 47656880) (47693948 47698103)del, g.47656881 47693947del,
g.(47643569 47656880) (47702410 47703505)del, g.47656881 47656882ins(173),
g.47656901 47656902insA, g.47656903de1, g.47656912de1, g.47630440de1,
g.47656923de1,
g.47656931 47656932dup, g.47656943 del, g.47656943 47656949delinsCCCAGA,
g.47656948dup, g.47656996dup, g.47657000 47657001dup, g.47630449de1,
g.47657007dup, g.47657008de1, g.47657020 47657023dup, g.47657025 47657026del,
g.47657026dup, g.47657030 4765703 ldel, g.47657047 47657050de1, g.47657053de1,

g.47657053 47657057del, g.47657064de1, g.47657073dup, g.47657312 47676594de1,
g.47668611 47674615del, g.47672116 47675123del, g.47666463 47677632del,
g.47666403 47677572del, g.(47657081 47672686) (47710367 ?)del,
g.(47657081 47672686) (47710367 ?)inv,
g.47671507 47675022delinsCATTCTCTTTGAAAA, g.47657278 47676557del,
g.47672687 47672796del, g.(47657081 47672686) (47672797 47690169)del,
g.(47657081 47672686) (47672797 47690169)del,
g.(47657081 47672686) (47693948 47698103)del,
g.(47657081 47672686) (47698202 47702163)del,
g.(47657081 47672686) (47708011 47709917)del, g.47672691dup, g.47672697dup,
g.47672721 47672744delins47672748 47672771inv, g.47672728 47672729del,
g.47672731dup, g.47672750 47672751insGG, g.47672755 47672758del,
g.47672762 47672763del, g.47630466 47630494del, g.47686194 47697740del,
g.(47672797 47690169) (47710367 ?)del,
g.(47672797 47690169) (47690294 47693796)del,
g.(47672797 47690169) (47693948 47698103)del, g.47690170 47693947del,
g.(47672797 47690169) (47693948 47698103)del,
g.(47672797 47690169) (47693948 47698103)dup,
g.(47672797 47690169) (47705659 47707834)del, g.47690173del, g.47690191del,
g.47690216 47690217dup, g.47690227del, g.47690227dup, g.47690228 47690232del,

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
g.47690230 4769023 ldel, g.47690240de1, g.47690240 47690243 del,
g.47630475de1,
g.47630475 47630476del, g.47690259 47690260delinsCT, g.47690277dup,
g.47690280del,
g.47690283dup, g.(47690294 47693796) (47702410 47703505)del,
g.47630484 47630485insG, g.47693838 47693839del, g.47693862del, g.47693864del,

g.47693873del, g.47693880dup, g.47693913del, g.47693924 47693925dup,
g.47630493del,
g.47697730 47706125del, g.(47693948 47698103) (47710367 ?)del,
g.(47693948 47698103) (47698202 47702163)del,
g.(47693948 47698103) (47705659 47707834)del, g.47698107del, g.47698109del,
g.47698109 47698110insA, g.47630496del, g.47698118del,
g.47698125del,g.47698129dup,
g.47698138 47698139del, g.47698142 47698146del, g.47698144dup,
g.47698147 47698148del, g.47698147 47698148dup, g.47698147 47698148insT,
g.47698159del, g.47698162del, g.47698506 47703472del, g.47701803 47708848del,
g.(47698202 47702163) (47710367 ?)del,
g.(47698202 47702163) (47702410 47703505)del,
g.(47698202 47702163) (47703711 47705410)del,
g.(47698202 47702163) (47705659 47707834)del, g.47702164del,
g.47702175 47702176insA, g.47702183 47702186del, g.47702185 47702186insCT,
g.47702190 47702192del, g.47702191dup, g.47702192 47702193del, g.47702213del,
g.47702231del, g.47702242dup, g.47702257del, g.47702262 47702263dup,
g.47630516 47630517dup, g.47630517del, g.47630517dup, g.47702289 47702290inv,
g.47702293 47702296del, g.47702301dup, g.47702315del, g.47702315del,
g.47702328 47702329del, g.47630522dup, g.47702339del, g.47702371 47702374dup,
g.47702384 47702385del, g.47702386 47702389del, g.47702388del,
g.47702388 47702389del, g.47702390del, g.47702390 47702391del,
g.47702400 47702401del, g.47703506 47703710del, g.47703506 47708010del,
g.47703510del, g.47703515del, g.47703521 47703522del, g.47703535 47703536del,
g.47703546 47703547del, g.47703548 47703611dup, g.47630534del, g.47703571dup,
g.47703574 47703581del, g.47703585dup, g.47630350del, g.47632107 47668733del,
g.47703613del, g.(47630542 47635539) (47643569 47656880)del,
g.(47630542 47635539) (47643569 47656880)inv,
g.(47630542 47635539) (47657081 47672686)del, g.47635540 47657080del,
g.(47630542 47635539) (47672797 47690169)del,
g.(47630542 47635539) (47690294 47693796)del,
66

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
g.(47630542 47635539) (47705659 47707834)del, g.47635540 47635694del,
g.(47630542 47635539) (47635695 47637232)del,
g.(47630542 47635539) (47635695 47637232)del,
g.(47630542 47635539) (47637512 47639552)del, g.47703635dup, g.47703641dup,
g.47635542 47635549del, g.47703660 47703663del, g.47703667dup, g.47630351dup,
g.47703704del, g.47703826 47707938del,
g.(47703711 47705410) (47705659 47707834)del, g.47705428 4770543 ldel,
g.47705437 47705438insA, g.47635551 47635552del, g.47705440 47705441del,
g.47705461del, g.47705490del, g.47705494del, g.47705495del, g.47635557
47635558del,
g.47705505del, g.47705535dup, g.47705547del, g.47705560 47705561dup,
g.47705561dup,
g.47705562dup, g.47705588del, g.47705608 47705609del, g.47705618dup,
g.47705627dup,
g.47635571 47635601delins(217), g.(47705659 47707834) (47710367 ?)del,
g.(47705659 47707834) (47708011 47709917)del, g.47707842 47707843del,
g.47707861del, g.47707861 47707874dup, g.47707878 47707884del,
g.47707878 47707884del, g.47707883 de!, g.47707895 47707905del, g.47707897del,

g.47707901 47707902del, g.47707905 47707906del, g.47707921del, g.47635583dup,
g.47635583 47635584del, g.47707969 47707973 de!, g.47707996 47707997ins(115),
g.47708009 47708010del, g.(47708011 47709917) (47710367 ?)del,
g.47635591 47635592del, g.47635597 47635618dup, g.47635606 47635607del,
g.47630359dup, g.47635672del, g.47635675 47635678del, g.47630364dup,
g.47635680dup,
g.47636862 47639040del, g.47636781 47638831del, g.47636753 47638155del,
g.47636552 47638597del, g.(47635695 47637232) (47643569 47656880)del,
g.(47635695 47637232) (47643569 47656880)del,
g.(47635695 47637232) (47657081 47672686)del,
g.(47635695 47637232) (47672797 47690169)del,
g.(47635695 47637232) (47698202 47702163)del,
g.(47635695 47637232) (47637512 47639552)del,
g.(47635695 47637232) (47641558 47643434)del, g.47637234del,
g.47637246 47637247del, g.47637253 47637254del, g.47637254 47637255del,
g.47637254 47637255del, g.47637265del, g.47637274del, g.47637282del,
g.47637320del,
g.47637372 47637375del, g.47637377 47637449dup, g.47637379del, g.47637384del,
g.47637394 47637395del, g.47637396 47637397del, g.47637417del,
g.47637427 47637435del, g.47637437 47637439del, g.47637453del, g.47637458dup,
67

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
g.47637479 47637482dup, g.47637482dup, g.47637504 47637505del,
g.47637508 47637511del, g.47638050 47653430de1, g.47638302 47648462del,
g.47638478 47648643del, g.(47637512 47639552) (47710367 ?)del,
g.(47637512 47639552) (47643569 47656880)del, g.47639553 47643568del,
g.(47637512 47639552) (47657081 47672686)del,
g.(47637512 47639552) (47657081 47672686)del,
g.(47637512 47639552) (47672797 47690169)del,
g.(47637512 47639552) (47639700 47641407)del,
g.(47637512 47639552) (47641558 47643434)del, g.47639557 47639561del,
g.47639582 47639586delinsTAAT, g.47639583 47639584de1, g.47639594de1,
g.47639594dup, g.47639598de1, g.47639603 47639604de1, g.47639611 47639612de1,
g.47639612de1, g.47639618 47639621del, g.47639624 47639628delinsTTA,
g.47630401dup, g.47639632dup, g.47639638 47639641dup, g.47639638 47639641dup,
g.47639639de1, g.47639639de1, g.47639642dup, g.47630403 47630404insC,
g.47639653del,
g.47639666de1, g.47639666 47639669de1, g.47639668de1, g.47639670
47639673delinsTT,
g.47639674 47639675dup, g.47639695 47639696de1, g.47639707 47642985de1,
g.47641402 47642007del, g.(47639700 47641407) (47643569 47656880)del,
g.47641408 47643568del, g.(47639700 47641407) (47657081 47672686)del,
g.(47639700 47641407) (47672797 47690169)del,
g.(47639700 47641407) (47641558 47643434)del,
g.(47639700 47641407) (47641558 47643434)del, g.47641410de1,
g.47641425 47641426del, g.47641426 47641429de1, g.47630412de1, g.47641451del,
g.47641454dup, g.47641455dup, g.47641469del, g.47641478del, g.47641488
47641491del,
g.47641496 47641497del, g.47641503de1, g.47641513 47641514dup,
g.47641530 47641537dup, g.47642509 47655432del,
g.(47641558 47643434) (47643569 47656880)del,
g.(47641558 47643434) (47693948 47698103)del, g.47630424 47630433del,
g.47643450dup, g.47643462 47643463del, g.47643462 47643463ins(4),
g.47643464 47643465insNC 000022.10:3578816935788352, g.47643465dup.
MutS Homolog 3 (MSH3) is a human homologue of the bacterial mismatch repair
protein MutS that participates in the mismatch repair (MMR) system. MSH3
typically forms
the heterodimer MutSP with MSH2 in order to correct long insertion/deletion
loops and base-
base mispairs in microsatellites during DNA synthesis. Deficient capacity for
MMR is found
68

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
in approximately 15% of colorectal cancers, and somatic mutations in the MSH3
gene can be
found in nearly 50% of MMR-deficient colorectal cancers. Examples of the
mutations in
MSH3 include, but are not limited to, g.79970809de1.
MSH6 encodes MutS homologue 6 (MSH6), a member of the Mutator S (MutS)
family of proteins that are involved in DNA mismatch repair (MMR). The MSH6
protein
forms a heterodimer with MutS homologue 2 (MSH2) in both human and yeast.
Human
MSH2/6 recognizes single base-base mismatches and short insertion/deletion
loops. Upon
recognition of a mismatch, MSH2/6 complex binds and exchanges ADP for ATP,
resulting in
a conformational change to the complex that precedes base pair dissolution,
base excision,
and repair.
MSH6 mutations include frameshift and/or nonsense mutations and can result in
non-
functional MSH6 and loss of protein expression. Examples include a frameshift
mutation at
MSH6 amino acid residue 290 and a compounding missense T1189I.
Inactivating MSH6 mutations can be detected in cancers by routine diagnostics
methods. These methods include, but are not limited to, obtaining cancer cells
and other
diagnostic indicators such as peripheral blood mononuclear cells (PBMCs), PBMC

subpopulations, circulating blasts (CD34+ cells), circulating tumor cells and
circulating
exosomescancer cells by biopsy and blood tests and by obtaining lymphatic or
other bodily
fluids. It is then determined from the cancer cells or other diagnostic
indicators whether the
cancer exhibits an inactivating MSH6 mutation is by methodology known in the
art, for
example, direct DNA sequencing and multiplex ligation dependent probe
amplification, RNA
sequencing (RNA-Seq), microarray, quantitative PCR, or NanoStrinem gene
expression
panels, or MSH6 protein by immunohistochemistry, flow cytometry,
immunocytochemistry
or Western blot. Methods for identifying inactivating MSH6 mutations are
disclosed in
Houlleberghs H, Goverde A, Lusseveld J, Dekker M, Bruno MJ, et al. (2017)
Suspected
Lynch syndrome associated MSH6 variants: A functional assay to determine their

pathogenicity. PLOS Genetics 13(5):
e1006765. https://doi.org/10.1371/journal.pgen.1006765.
Examples of the mutations in MSH6 include, but are not limited to,
g.48032846 48032849del, g.48032846 48032849de1, g.48032846 48032849del,
g.48033337 48033342del, g.48033420 48033422de1, g.(? 48010221) (48034092)del,
g.(? 48010221) (48018263 48023032)del, g.47998510 48020183del,
g.48007276 48020272del, g.48026207de1, g.48026223 del, g.48026223 del,
69

CA 03133005 2021-09-08
WO 2020/198298
PCT/US2020/024601
g.48026257 48026261del, g.48026261 48026265de1, g.48026312 48026313del,
g.48026398de1, g.48026543 48026544dup, g.48026693dup, g.48026702de1,
g.48026712de1,
g.48026718dup, g.48026736 48026737delinsAG, g.48026736 48026737delinsG,
g.48026750 48026751del, g.48026754 48026757de1, g.48026756 48026759del,
g.48026759 48026760del, g.48026906de1, g.48026928 4802693 ldel, g.48026941dup,

g.48026991del, g.48027023 48027024del, g.48027079del, g.48027079 48027082dup,
g.48027167 48027168del, g.48027172 48027173dup, g.48027178 48027185del,
g.48027184 48027185del, g.48027272 48027275del, g.48027470 48027471del,
g.48027501 48027502del, g.48027501 48027502delTG, g.48027657dup,
g.48027691 48027694del, g.48027733 48027736dup, g.48027794 48027796delinsC,
g.48027841 48027842del, g.48027887del, g.48027890dup, g.48027973 48027980del,
g.48028067del, g.48028098del, g.48028106del, g.48028175 48028176del,
g.48028241 48028242del, g.48028241 48028242delTT, g.48028272 48028284dup,
g.48028277 48028278del, g.48030558 48030559del, g.48030126 48032394del,
g.48030568del, g.48030581 48030584del, g.48030584 48030585dup, g.48030607del,
g.48030645 48030646insT, g.48030647del, g.48030647dup, g.48030649dup,
g.48030654 48030660del, g.48030659dup, g.48030697 48030698del, g.48030698del,
g.48030706del, g.48030710dup, g.48030727 48030728insC, g.48030765 48030829del,

c.3438+797 3438+798insTATins1839 3439-428,
c.3438+797 3438+798insTATins1839 3439-428, g.48032121 48032122del,
g.48032123 48032124del, g.48032124dup, g.48032126 48032129del,
g.48032129 48032130insA, g.48032129 48032132dup,
g.(48032167 48032756) (48034092 ?)del, g.48032809 48032812del, g.48032835dup,
g.48032846 48032849del, g.48033374 48033402dup, g.48033395 48033398del,
g.48033421 48033433del, g.48033425 48033428dup, g.48033453 48033454insA,
g.48033494 48033523del, g.48033495 48033496del, g.48033593dup,
g.48033610 48033613dup, g.48033629 48033635del, g.48033636 48033639dup,
g.48033676 48033682del, g.48033707dup, g.48033709 48033716dup,
g.48033721 48033724dup, g.48033727 48033730dup, g.48033728 48033746dup,
g.(48033742 48033743) (48033742 48033743)ins(32), g.48033746dup,
g.48033748 48033751del, g.48033758 48033768del, g.48033773 48033774insATCA,
g.48033773 48033776dup, g.48033785 48033789dup, g.48033887 48033910inv,
g.(48018263 48023032) (48032167 48032756)del,

CA 03133005 2021-09-08
WO 2020/198298
PCT/US2020/024601
g.(48018263 48023032) (48023203 48025749)del, g.48023097 48023098del,
g.48025773dup, g.48025832de1, g.48025860 48025861insT, g.48025884 48025885del,

g.48025967dup.
MutL homolog 1, colon cancer, nonpolyposis type 2 (E. coli) is a protein that
in
humans is encoded by the MLH1 gene located on Chromosome 3. It is a gene
commonly
associated with hereditary nonpolyposis colorectal cancer.
Examples of the mutations in MSH6 include, but are not limited to,
g.37089113 37089115del, g.37089175de1, g.37090379 37090393del,
g.37038201 37038202del, g.37042531 37042542de1, g.37053339 37053355del,
g.37053354de1, g.37053590 37053591insT, g.37034841 37092337de1,
g.(? 37034841) (37092337 ?)del, g.(? 37034841) (37061955 37067127)del,
g.(? 37034841) (37035155 37038109)del, g.(? 37034841) (37035155 37038109)del,
g.(? 37034841) (37070424 37081676)del, g.(? 37034841) (37083823 37089009)del,
g.37034841 37083822de1, g.(? 37034841) (37038201 37042445)del,
g.(? 37034841) (37042545 37045891)del, g.37034841 37042544de1,
g.(? 37034841) (37042545 37045891)del, g.(? 37034841) (37042545 37045891)del,
g.(? 37034841) (37045966 37048481)del, g.(? 37034841) (37050397 37053310)del,
g.(? 37034841) (37059091 37061800)del, g.37034658 37038806del,
g.36961079 37138741del, g.37061923del, g.37061927del, g.37061933del,
g.37061939del,
g.37061942dup, g.37035140 37035141del, g.37070417de1, g.37070417 37070418insT,

g.37070419dup, g.37070422 37070423insT, g.37080355 37083368de1,
g.(37070424 37081676) (37092337 ?)del,
g.(37070424 37081676) (37081786 37083758)del,
g.(37070424 37081676) (37083823 37089009)del, g.37038148 37038151del,
g.37038149de1, g.37038149dup, g.37081690 37081691de1, g.37081691 37081692del,
g.37081706 37081708del, g.37081710 37081711de1, g.37035053 37035066del,
g.37038154de1, g.37038154 37038157de1, g.37081738 37081739de1, g.37081740de1,
g.37081753dup, g.37081757 37081761dup, g.37081782 37081783insAAGT,
g.37081787 37081793delinsATTT, g.(37081786 37083758) (37083823 37089009)del,
g.(37081786 37083758) (37089175 37090007)del, g.37083759de1, g.37083780dup,
g.37083781 37083784de1, g.37083781 37083784delCTCA, g.37083808 37083809del,
g.37083816del, g.37086069 37089606de1, g.37084092 37089247del,
g.37084590 37089786del, g.(37083823 37089009) (37092337 ?)del,
71

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
g.(37083823 37089009) (37089175 37090007)del, g.37089010 37089174del,
g.(37083823 37089009) (37090509 37091976)del, g.37089023del,
g.37089026 37089027del, g.37089027de1, g.37089036de1, g.37089036dup,
g.37038168dup,
g.37089042de1, g.37089047de1, g.37089050 37089053de1, g.37089056 37089057del,
g.37089061 37089062del, g.37089078 37089096de1, g.37089090dup, g.37089099dup,
g.37089107 37089110dup, g.37089109 37089110del, g.37089130 37089132del,
g.37089130 37089132delAAG, g.37089131delinsTTCTT, g.37089133de1,
g.37089133delG,
g.37089144de1, g.37089155de1, g.37089155 37089161de1, g.37089158 37089161del,
g.37089162 37089166de1, g.37089171del,
g.(37089175 37090007) (37090101 37090394)del, g.37035056 37035072de1,
g.37090013de1, g.37090015dup, g.37038183 37038184de1, g.37090024 37090037dup,
g.37090025 37090053dup, g.37090027dup, g.37038184dup, g.37090031 37090032insT,

g.37090041del, g.37090057de1, g.37090064 37090067de1, g.37038188de1,
g.37090082de1,
g.37090086 37090087del, g.37090087 37090088de1, g.37090097 37090101delinsC,
g.37090099de1, g.37038191dup, g.(3709010137090394) (37092337 ?)del,
g.37035057 37035073del, g.37090405dup, g.37090411 37090415del, g.37090414de1,
g.37038194de1, g.37038198de1, g.37090472 37090478de1, g.37039445 37059613dup,
g.37039760 37052440del, g.37090481 37090482del, g.37090483 37090484del,
g.37090483 37092045de1, g.37040732 37043185delinsACATAGTA,
g.37042445 37042446del, g.(3703820137042445) (37042545 37045891)del,
g.(3703820137042445) (37048555 37050304)del,
g.(3703820137042445) (37050397 37053310)del,
g.(37038201 37042445) (37053591 37055922)del, g.37090497 37090498del,
g.37090497 37090498delTC, g.37090504 37090507del,
g.(37090509 37091976) (37092337 ?)del, g.(37090509 37091976) (37092337 ?)dup,
g.37091977 37091978del, g.37091978 37091987de1, g.37042448 37042451del,
g.37091984 37091990del, g.37042451 37042453de1, g.37092020 37092021del,
g.37092022 37092068dup, g.37092027 37092028de1, g.37092027 37092028dup,
g.37092030dup, g.37092052 37092055de1, g.37092054 37092055del,
g.37092068 37092071dup, g.37092091dup, g.37092094 37092097delins(30),
g.37092096 37092106del, g.37092097de1, g.37092125 37092126delAA,
g.37092125 37092126de1, g.37092139 37092142dup, g.37092142dup, g.37035060dup,
g.37042469 37042470del, g.37042470de1, g.37042482dup, g.37042485de1,
g.37042499de1,
72

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
g.37042546dup, g.37044472 37046589del, g.37045648 37049941del,
g.37045095 37054651del, g.37045072 37046861del,
g.(37042545 37045891) (37045966 37048481)del,
g.(37042545 37045891) (37092337 ?)del,
g.(37042545 37045891) (37048555 37050304)del,
g.(37042545 37045891) (37050397 37053310)del, g.37045892 37050396del,
g.37035069del, g.37045926del, g.37045931del, g.37045939 37045940dup,
g.37045957 37045958del, g.37045963del, g.37035075del, g.37048067 37049287del,
g.(37045966 37048481) (37048555 37050304)del,
g.(37045966 37048481) (37050397 37053310)del, g.37048483del,
g.37048483 37048503delinsT, g.37048486 37048487delinsGTT, g.37048489del,
g.37048490del, g.37035076 37035077insCCCA, g.37035077 37035078dup,
g.37048505 37048508del, g.37048521del, g.37048529dup, g.37035082dup,
g.37049873 37052281del, g.37049839 37052249del, g.37049800 37052209del,
g.37049640 37050445del, g.37050305 37050396del,
g.(37048555 37050304) (37050397 37053310)del, g.37050305 37050396del,
g.37050319 37050320del, g.37050339del, g.37050348del, g.37050353 37050354del,
g.37050354dup, g.37050364del, g.37050375 37050376insGA, g.37035090del,
g.37050382 37050383delinsAT, g.37050382 37050383delinsCT, g.37050390
37050396del,
g.37052950 37060990del, g.(37050397 37053310) (37067499 37070274)dup,
g.(37050397 37053310) (37053591 37055922)del,
g.(37050397 37053310) (37056036 37058996)del, g.37053353del,
g.37053510 37053511del, g.37035099del, g.37053545 37053546insT, g.37053562del,
g.37053578del, g.37053578dup, g.37053585del, g.37053586 37053589del,
g.37053591del,
g.37053590 37053591delinsAT, g.37055920 37055921del, g.37055914 37055938del,
g.(37053591 37055922) (37070424 37081676)del,
g.(37053591 37055922) (37083823 37089009)del,
g.(37053591 37055922) (37059091 37061800)del, g.37035105del, g.37055928dup,
g.37035106 37035116del, g.37055938del, g.37035108del, g.37055972 37055975del,
g.37055976 37055979del, g.37035111del, g.37055990dup, g.37035114del,
g.37035116del,
g.37056036del, g.37056037dup, g.37058993 37059001del,
g.(37056036 37058996) (37070424 37081676)del,
g.(37056036 37058996) (37059091 37061800)del, g.37058997 37059000del,
73

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
g.37059014 37059017del, g.37059017 37059021del, g.37059027 37059030dup,
g.37035122del, g.37059062 37059063insT, g.37059065 37059066del, g.37059066del,

g.37059066dup, g.37059072 37059073del, g.37059072 37059073dup,
g.37059090 37059093del, g.37061595 37061913del, g.37061308 37066756del,
g.37061207 37063077del, g.(37059091 37061800) (37092337 ?)del,
g.(37059091 37061800) (37061955 37067127)del, g.37061801 37061954del,
g.(37059091 37061800) (37083823 37089009)del, g.37061803dup, g.37061804del,
g.37061817del, g.37061837 37061838dup, g.37061844del, g.37061851dup,
g.37061855dup,
g.37061870del, g.37061904 37061906del, g.37061910del, g.37035047del,
g.[37049179 37051317delinsTG;37051667 37054327delinsCA].
Human PMS2 related genes are located at bands 7p12, 7p13, 7q11, and 7q22.
Exons 1
through 5 of these homologues share high degree of identity to human PMS2. The
product of
this gene is involved in DNA mismatch repair. The protein forms a heterodimer
with MLH1
and this complex interacts with MSH2 bound to mismatched bases. Defects in
this gene are
associated with hereditary nonpolyposis colorectal cancer, with Turcot
syndrome, and are a
cause of supratentorial primitive neuroectodermal tumors.
Examples of the mutations in PMS2 include, but are not limited to,
g.(? 6012870) (6048737 ?)del, g.6012870 6048737del,
g.(6027252 6029430) (6048737 ?)del, g.(6045663 6048627) (6048737 ?)del,
g.6029554del, g.6029499dup, g.6029495 6029496del, g.6029462 6029463delinsTAAA,

g.5992485 6028601del, g.(6018328 6022454) (6027252 6029430)del,
g.(6013174 6017218) (6027252 6029430)del, g.6027226 6027227in5(20),
g.6027175del,
g.6027090dup, g.6036705 6044207delinsCG, g.6026666dup, g.6026628del,
g.6043671del,
g.6026565dup, g.6026565dupT, g.6018315 6018316del, g.6018306 6018310del,
g.6018306 6018310delAGTTA, g.6043633 6043634dup, g.6018256 6018259del,
g.6015623 6017501del, g.6016429 6017479del, g.6017300 6017303del,
g.6045579 6045674delinsATTT, g.(6043690 6045522) (6045663 6048627)del,
g.(? 6012870) (6042268 6043320)del, g.(6035265 6036956) (6042268 6043320)del,
g.6038283 6039384del, g.6038901del, g.6038851dup,
g.(6035265 6036956) (6037055 6038738)del,
g.6037019 6037024delinsCTTCACACACA, g.6036980del, g.6036958dup,
g.6035323 6035324insJN866832.1, g.(6022623 6026389) (6035265 6036956)del,
g.(6031689 6035164) (6035265 6036956)del, g.6035204 6035207del,
74

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
g.6035205 6035206de1, g.(? 6012870) (6031689 6035164)del,
g.(6027252 6029430) (6031689 6035164)del,
g.(6029587 6031603) (6031689 6035164)del, g.6028725 6029882del,
g.(? 6012870) (6029587 6031603)del.
The present application provides a method of treating patients with Lynch
syndrome
to reduce the likelihood of from developing or treating cancers derieved from
Lynch
syndrome, by administering to the subject a therapeutically effective amount
of one or more
disclosed compounds, or a pharmaceutically acceptable salt thereof, or the
corresponding
pharmaceutical composition in combination with a PARP inhibitor or a
pharmaceutically
acceptable salt thereof or a pharmaceutical composition comprising a PARP
inhibitor.
Lynch syndrome is a hereditary disorder caused by a mutation in a mismatch
repair
gene in which affected individuals have a higher than normal chance of
developing colorectal
cancer, endometrial cancer, and various other types of aggressive cancers,
often at a young
age ¨ also called hereditary nonpolyposis colon cancer (HNPCC).
The mutations of specific mismatch repair (MMR) genes including but not
limited to
MLH1, MSH2, MSH6, PMS2, and EPCAM-TACSTD1 deletions are responsible for Lynch
syndrome. These genes work in repairing mistakes made when DNA is copied in
preparation
for cell division. The defects in the genes disallow repair of DNA mistakes
and as cells
divide, errors stack and uncontrollable cell growth may result in cancer.
Those with Lynch syndrome carry up to an 85% risk of contracting colon cancer
as
well as a higher than average risk for endometrial cancer, stomach cancer,
pancreatic cancer,
kidney/ureter tract cancer, hepatobiliary tract cancer, gastric tract cancer,
prostate cancer,
ovarian cancer, gallbladder duct cancer, brain cancer, small intestine cancer,
breast cancer,
and skin cancer.
Thus, in one embodiment for the disclosed method, the method is a method of
treating
cancer derived from Lynch syndrome, selected from the group consisting of
colon cancer,
endometrial cancer, stomach cancer, pancreatic cancer, kidney/ureter tract
cancer,
hepatobiliary tract cancer, gastric tract cancer, prostate cancer, ovarian
cancer, gallbladder
duct cancer, brain cancer, small intestine cancer, breast cancer, and skin
cancer.
In yet another embodiment, the method is a method of treating autoimmune
disease.
Exemplary autoimmune diseases include lupus erythematosus; Wiskott-Aldrich
syndrome;
autoimmune lymphoproliferative syndrome; myasthenia gravis; rheumatoid
arthritis (RA);
lupus nephritis; multiple sclerosis; systemic lupus erythematosis; discoid
lupus; subacute

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
cutaneous lupus erythematosus; cutaneous lupus erythematosus including
chilblain lupus
erythematosus; chronic arthritis; Sjogren's syndrome; inflammatory chronic
rhinosinusitis;
colitis; celiac disease; inflammatory bowel disease; Barrett's esophagus;
inflammatory
gastritis; autoimmune nephritis; autoimmune vasculitis; autoimmune hepatitis;
autoimmune
carditis; autoimmune encephalitis; autoimmune diabetes; autoimmune diabetes
nephritis;
psoriasis; Graft-versus-host disease (GvHD); and autoimmune mediated
hematological
disease.
In one aspect of this embodiment, the method is a method of treating immune
deficiency selected from the group consisting of Autoimmune
Lymphoproliferative
Syndrome (ALPS), Autoimmune polyglandular syndrome type 1 (APS-1), BENTA
Disease,
Caspase Eight Deficiency State (CEDS), Chronic Granulomatous Disease (CGD),
Common
Variable Immunodeficiency (CVID), Congenital Neutropenia Syndromes, CTLA4
Deficiency, DOCK8 Deficiency, GATA2 Deficiency, Glycosylation Disorders With
Immunodeficiency, hyper-immunoglobulin E syndrome (HIES), Hyper-Immunoglobulin
M
(Hyper-IgM) Syndromes, Leukocyte adhesion deficiency (LAD), LRBA deficiency,
PI3
Kinase disease, PLCG2-associated antibody deficiency and immune dysregulation
(PLAID),
severe combined immunodeficiency (SCID), STAT3 gain-of-function disease,
Warts,
Hypogammaglobulinemia, Infections, and Myelokathexis Syndrome (WHIMS), X-
Linked
Agammaglobulinemia (XLA), X-Linked Lymphoproliferative Disease (XLP), and XMEN

Disease.
As used herein, the term "immune deficiency" refers to a condition in which a
portion
or some portions of cell components constituting an immune system are
defective or
dysfunction, so that a normal immune mechanism is damaged. In other words,
"immune
deficiency" means a condition under which: congenital immunity and/or acquired
immunity
are suppressed and/or decreased. In some embodiments, the immune -deficiency
subject is an
immunocompromised subject. Non-limiting examples of immune deficiencies can
include
AIDS, hypogammaglobulinemia, agammaglobulinemia, granulocyte deficiency,
chronic
granulomatous disease, asplenia, SCID, complement deficiency, and/or sickle
cell anemia.
In another aspect of this embodiment, the method is a method of treating a
neurodegenerative disorder selected from the group consisting of multiple
sclerosis,
Parkinson's disease (PD), Alzheimer's disease (AD), Dentatorubropallidoluysian
atrophy
(DRPLA), Huntington's Disease (HD), Spinocerebellar ataxia Type 1 (SCA1),
Spinocerebellar ataxia Type 2 (SCA2), Spinocerebellar ataxia Type 3 (SCA3),
76

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
Spinocerebellar ataxia 6 (SCA6), Spinocerebellar ataxia Type 7 (SCA7),
Spinocerebellar
ataxia Type 8 (SCA8), Spinocerebellar ataxia Type 12 (SCA12), Spinocerebellar
ataxia Type
17 (SCA17), Spinobulbar Muscular Ataxia/Kennedy Disease (SBMA), Fargile X
syndrome
(FRAXA), Fragile XE mental retardation (FRAXE), and Myotonic dystrophy (DM).
A "subject" is a mammal, preferably a human, but can also be an animal in need
of
veterinary treatment, e.g., companion animals (e.g., dogs, cats, and the
like), farm animals
(e.g., cows, sheep, pigs, horses, and the like) and laboratory animals (e.g.,
rats, mice, guinea
pigs, and the like).
In some embodiments, the methods disclosed herein further comprise co-
administering a therapeutically effective amount of a DNA repair inhibitor, a
DNA damage
response (DDR) inhibitor, a DNA damaging agent or an immunomodulatory agent to
the
subject being treated for cancer, in addition to a therapeutically effective
amount of a
disclosed RAD51 inhibitor.
The term "DNA repair inhibitor" refers to any agent that targets
components/processes which a cell uses to repair mutations or changes in DNA
and restore
the DNA to its original state and prevents the repair of DNA. Examples of DNA
repair
inhibitors include: RPA inhibitors, APE1 inhibitors, DNA ligase inhibitors,
DNA polymerase
inhibitors, Parp inhibitors etc.
The term "DNA damage response inhibitor" refers to any agent that targets
components/processes involved in detecting DNA lesions, signaling the presence
of DNA
damage, and/or promote the repair of DNA damage. Examples of DNA damage
response
inhibitors include checkpoint inhibitors, ATM and ATR inhibitiors, DNA-PK
inhibitors, etc.
The term "DNA damaging agent" refers to any agent that directly or indirectly
damages DNA for which homologous recombination could repair the damage. The
DNA
damaging agents is selected from the group consisting of: exposure to a DNA
damaging
chemical; exposure to a chemotherapeutic agent; exposure to a
radiochemotherapy, and
exposure to ionizing or ultraviolet radiation. Specific examples of DNA-
damaging
chemotherapeutic agents include alkylating agents, nitrosoureas, anti-
metabolites, plant
alkaloids, plant extracts and radioisotopes. Specific examples of the
chemotherapeutic agents
also include DNA-damaging drugs, for example, 5-fluorouracil (5-FU),
capecitabine, 5-1
(Tegafur, 5-chloro-2,4-dihydroxypyridine and oxonic acid), 5-ethynyluracil,
arabinosyl
cytosine (ara-C), 5-azacytidine (5-AC), 2',2'-difluoro-2'-deoxycytidine
(dFdC), purine
antimetabolites (mercaptopurine, azathiopurine, thioguanine), gemcitabine
hydrochlorine
77

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
(Gemzar), pentostatin, allopurinol, 2-fluoro-arabinosyl-adenine (2F-ara-A),
hydroxyurea,
sulfur mustard (bischloroetyhylsulfide), mechlorethamine, melphalan,
chlorambucil,
cyclophosphamide, ifosfamide, thiotepa, AZQ, mitomycin C, dianhydrogalactitol,

dibromoducitol, alkyl sulfonate (busulfan), nitrosoureas (BCNU, CCNU, 4-methyl
CCNU or
ACNU), procarbazine, decarbazine, rebeccamycin, anthracyclins such as
doxorubicin
(adriamycin; ADR), daunorubicin (Cerubicine), idarubicin (Idamycin) and
epirubicin
(Ellence), anthracyclin analogs such as mitoxantrone, actinimycin D, non-
intercalating
topoisomerase inhibitors such as epipodophyllotoxins (etoposide or VP16,
teniposide or VM-
26), podophylotoxin, bleomycin (Bleo), pepleomycin, compounds that form
adducts with
nucleic acid including platinum derivatives, e.g., cisplatin (CDDP), trans
analog of cisplatin,
carboplatin, iproplatin, tetraplatin and oxaliplatin, as well as camptothecin,
topotecan,
irinotecan (CPT-11), and SN-38. Specific examples of nucleic acid damaging
treatments
include radiation e.g., ultraviolet (UV), infrared (IR), or .alpha.-, .beta.-,
or .gamma.-
radiation, as well as environmental shock, e.g., hyperthermia.
"Immunomodulatory agent" means an agent that modulates an immune response to
an
antigen but is not the antigen or derived from the antigen. "Modulate", as
used herein, refers
to inducing, enhancing, suppressing, directing, or redirecting an immune
response. Such
agents include immunostimulatory agents, such as adjuvants, that stimulate (or
boost) an
immune response to an antigen but is not an antigen or derived from an
antigen. There are
several distinct types of immunomodulatory agents, which include, but are not
limited to,
Toll-like Receptor (TLR) agonists and Toll-like Receptor (TLR) antagonists.
Such agents
also include immunosuppressants. The immunomodulatory agent is selected from
the group
consisting of immune checkpoint modulators, Toll-like receptor (TLR) agonists,
cell-based
therapies, cytokines and cancer vaccines.
In some embodiments, the subject is determined to have an increased level
and/or
activity of a DNA damage process or DNA editing enzyme. In one aspect of this
embodiment, the DNA editing enzyme is selected from the group consisting of
activation
induced cytidine deaminase (AID or AICDA), APOBEC2, APOBEC3A, APOBEC3C,
APOBEC3D, APOBEC3F, APOBEC3G, APOBEC3H, APOBEC4, a Type 1 Topoisomerase,
a Type 2 Topoisomerase, Recombination Activating Gene 1 (RAG 1), and
Recombination
Activating Gene 2 (RAG2).
In some embodiments, blood cells obtained from the subject have been
determined to
have a detectable level of activation-induced cytidine deaminase (AID).
78

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
In some embodiments, B cells obtained from the subject have been determined to

have a detectable level of activation-induced cytidine deaminase (AID).
In some embodiments, the detectable level of activation-induced cytidine
deaminase
(AID) is statistically significantly higher than the level of AID expressed in
unactivated B-
cells or normal non-immune cells from a healthy subject.
Methods of Administration and Dosage Forms
The precise amount of compound administered to provide an "effective amount"
to
the subject will depend on the mode of administration, the type, and severity
of the disease,
and on the characteristics of the subject, such as general health, age, sex,
body weight, and
tolerance to drugs. The skilled artisan will be able to determine appropriate
dosages
depending on these and other factors. When administered in combination with
other
therapeutic agents, e.g., when administered in combination with an anti-cancer
agent, an
"effective amount" of any additional therapeutic agent(s) will depend on the
type of drug
used. Suitable dosages are known for approved therapeutic agents and can be
adjusted by the
skilled artisan according to the condition of the subject, the type of
condition(s) being treated
and the amount of a compound of the application and a PARP inhibitor being
used by
following, for example, dosages reported in the literature and recommended in
the
Physician's Desk Reference (57th ed., 2003).
The term "effective amount" means an amount when administered to the subject
which results in beneficial or desired results, including clinical results,
e.g., inhibits,
suppresses or reduces the symptoms of the condition being treated in the
subject as compared
to a control. For example, a therapeutically effective amount can be given in
unit dosage form
(e.g., 0.1 mg to about 50 g per day, alternatively from 1 mg to about 5 grams
per day).
The terms "administer", "administering", "administration", and the like, as
used
herein, refer to methods that may be used to enable delivery of compositions
to the desired
site of biological action. These methods include, but are not limited to,
intraarticular (in the
joints), intravenous, intramuscular, intratumoral, intradermal,
intraperitoneal, subcutaneous,
orally, topically, intrathecally, inhalationally, transdermally, rectally, and
the like.
Administration techniques that can be employed with the agents and methods
described
herein are found in e.g., Goodman and Gilman, The Pharmacological Basis of
Therapeutics,
current ed.; Pergamon; and Remington's, Pharmaceutical Sciences (current
edition), Mack
Publishing Co., Easton, Pa.
79

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
The particular mode of administration and the dosage regimen will be selected
by the
attending clinician, taking into account the particulars of the case (e.g.,
the subject, the
disease, the disease state involved, the particular treatment). Treatment can
involve daily or
multi-daily or less than daily (such as weekly or monthly etc.) doses over a
period of a few
days to months, or even years. However, a person of ordinary skill in the art
would
immediately recognize appropriate and/or equivalent doses looking at dosages
of approved
compositions for treating a a RAD51 mediated disease using the disclosed RAD51
inhibitors
for guidance.
The compounds or the corresponding pharmaceutical compositions taught herein
and
the PARP inhibitor or a pharmaceutically acceptable salt thereof or a
pharmaceutical
composition comprising a PARP inhibitor can be administered to a patient in a
variety of
forms depending on the selected route of administration, as will be understood
by those
skilled in the art. The compounds of the present application and the PARP
inhibitors may be
administered, for example, by oral, parenteral, buccal, sublingual, nasal,
rectal, patch, pump
or transdermal administration and the pharmaceutical compositions formulated
accordingly.
Parenteral administration includes intravenous, intraperitoneal, subcutaneous,
intramuscular,
transepithelial, nasal, intrapulmonary, intrathecal, rectal and topical modes
of administration.
Parenteral administration can be by continuous infusion over a selected period
of time.
The pharmaceutical composition of the application is formulated to be
compatible
with its intended route of administration. In some embodiments, the
composition is
formulated in accordance with routine procedures as a pharmaceutical
composition adapted
for intravenous, subcutaneous, intramuscular, oral, intranasal, or topical
administration to
human beings. In some embodiments, the pharmaceutical composition is
formulated for
intravenous administration.
In some embodiments for oral therapeutic administration, a compound of the
present
application and the PARP inhibitors may be incorporated with excipient and
used in the form
of ingestible tablets, buccal tablets, troches, capsules, elixirs,
suspensions, syrups, wafers, and
the like.
In some imbodiments for parenteral administration, solutions of a compound of
the
present application and the PARP inhibitors can generally be prepared in water
suitably
mixed with a surfactant such as hydroxypropylcellulose. Dispersions can also
be prepared in
glycerol, liquid polyethylene glycols, DMSO and mixtures thereof with or
without alcohol,

CA 03133005 2021-09-08
WO 2020/198298
PCT/US2020/024601
and in oils. Under ordinary conditions of storage and use, these preparations
contain a
preservative to prevent the growth of microorganisms.
In some embodiments for injectable use, sterile aqueous solutions or
dispersion of,
and sterile powders of, a compound described herein for the extemporaneous
preparation of
sterile injectable solutions or dispersions are appropriate.
EXAMPLES
Abbreviations:
Ac acetyl
ACN acetonitrile
aq aqueous
Bn benzyl
Boc tert-butoxycarbonyl
br. broad
doublet (only when used within 1H NMR spectra)
DCM dichloromethane
DIEA(DIPEA) diisopropylethylamine
DMA dimethylacetamide
DMAP 4-dimethylaminopyridine
DMF N,N-dimethylformamide
DMSO dimethylsulfoxide
dppf 1,1'- bis( diphenylphosphino) ferrocene
eq equivalent
Et0Ac ethyl acetate
hr hour
HBTU N,N,N',N',-tetramethy1-0-(1H-benzotriazol-1-y1)uronium
hexafluorophosphate
HPLC high performance liquid chromatography
LC-MS liquid chromatography coupled to mass spectrometry
multiplet
MS ESI mass spectra, electrospray ionization
NB S N-bromosuccinimide
NMR nuclear magnetic resonance
81

CA 03133005 2021-09-08
WO 2020/198298
PCT/US2020/024601
prep preparative
Py pyridine
singlet
sat saturated
SFC supercritical fluid chromatography
triplet
TEA triethylamine
TFA trifluoroacetic acid
THF tetrahydrofuran
TLC thin layer chromatography
Tol toluene
General Method Reaction Name
A Substitution Reaction
Suzuki Reaction A
Deprotection of Boc group A (TFA)
Acylation Reaction
Urea Formation
Deprotection of Boc group B (HCl)
Reduction with Fe
Carbamate Formation
Hydrogenation
Bromination
Suzuki Reaction B
Thionation
Cyclization
Coupling Reaction
0 Hydrolysis Reaction
Example 1. Synthesis of Compounds Compound 67A and Compound 67B
Compounds of the present application can be prepared according to the schemes,

methods, and examples described in US 20190077799. The procedures below
describe the
synthesis of compounds Compound 67A and Compound 67B.
82

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
-....?
, N N
4. / \__ 0
¨cA th ,o
/ %,_ i7.-B
W
-Br
Jet-, -Br
NHBoc
H2N 0 _____________________________________________________ v.
Py, DCM H S Pd(dppf)C12, Na2CO3,
/ '0
dioxane/H20, 80 C
35 42
0 NHBoc 0 0 N \ N
¨ (3,--1(N 40 S 11111 Pd/C, H DAN 40 S o
NHBoc _ii..HCl/Me0H
i.... ,
H ,
S( Me0H H S(
NH
43 "
/ \ 0 N
/ \ 0
CAN 40 -oS)-0-"NH2 LC31C1 *I(N 440 S N)-----(i)-- X. ).----- SF0
,0
H H
Py, DCM N H
.....)_.-NH
45 46
N
____c_jO( 40 is......0 Ox )..,.,,
---Cl(N lk

N '"N
,0 +
H H H S. H
/ 0 -0
....)--NH ....)--NH/
Ex. 67A Ex. 67B
Preparation of compound 42
, N N
Br
-Br )L OAN S
a o
S Py, DCM H S
/ (:) i (:)
35 42
General method D, isopropyl N-1-4-(2-bromothiazol-5-yl) -3-(tert-
butylsulfamoyl)
phenyl] carbamate. ESI [M+H] =476.0/478Ø
Preparation of compound 43
----ci)
N B alb N
4110 / o'
/ \
Br W i 0
--M-A
S 17 NHBoc D N H H S 4111 s
0 (
,0 NHBoc
/ '0 Pd(dppf)C12, Na2CO3, 1.
dioxane/H20, 80 C
42 43
General method B, isopropyl N-[4-[2-[4-(tert -butoxycarbonylamino)cyclohexen-l-
yl]
thiazol-5-yl]-3-(tert-buOsulfamoyl)phenyl] carbamate . ESI [M+H] =593.3.
Preparation of compound 44
83

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
i o
---- o--1.(N N
/ \
S 6 ,0 N
H 0
NHBoc Pd/C, El 3,... ¨ -1 --1( H N 1=/S)--0--NHBoc
S
Me0H
43 44
General method I, isopropyl N-1-4-12-1-4-(tert-butoxycarbonylamino)cyclohexyli
thiazol-5-y1]-
3-(tert-butylsulfamoyOphenylicarbamate. ESI [M+H] =595.3.
Preparation of compound 45
i o
/ \
NHBoc HCl/Me0H,
/ \
H 0
H 0
NH2
N'0
.....)._-NH H
44 45
General method F, isopropyl N-1-4-[2-(4-aminocyclohexyl) thiazol-5-y1]-3-(tert-

butylsulfamoyOphenylicarbamate. ESI [M+H] =495.2.
Preparation of compound 46
I o o
0
)---.0)-----
COAN 10 -/oS)0,N NH2 =joici N N
,0
H S H S H
i '0 / '0
... Py, DCM
45 46
General method D, isopropyl N-0-(tert-butylsulfamoy1)-4-12-1-4-
(isopropoxycarbonylamino)
cyclohexylithiazol-5-yliphenylicarbamate. ESI [M+H] =581.2.
Preparation of Compound 67A and Compound 67B.
o I \ o I N\I 0
--CAN * -oSraN NX-0)--- SFC ----c-A N 0 -oSr0 )--- )-----
'"N H
46 Ex. 67A
, N
+
---"CiZH *

H
Ex. 67B
Compound 46 was separated by SFC (Instrument: Thar SFC80 preparative SFC;
Column: ChiralpakAD-H 250*30mm i.d. 5u; Mobile phase: A for CO2 and B for
IPA(0.1%NH3H20); Gradient: B%=30%; Flow rate:70 g/min; Wavelength:220 nm;
Column
temperature: 40 C; System back pressure: 100 bar; Cycle time:8 min; Injection
amount: 3 mg
per injection); and then purified by prep-HPLC (Column: Agela Durashell C18
150*25 5u;
84

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
mobile phase: [water(0.1%TFA)-ACN];B%: 55%-85%,12min), trans-isopropylN-1-3-
(tert-
butylsulfamoyl)-4-12-14-(isopropoxycarbonylamino)cyclohexylithiazol-5-
yliphenylicarbamate Compound 67A (5.76 mg, 100% purity) and cis-isopropylN-1-3-
(tert-
butylsulfamoyl)-4-12-14-(isopropoxycarbonylamino) cyclohexylithiazol-5-
yliphenylicarbamate Compound 67B (3.95 mg, 100% purity) were obtained as a
pale yellow
solid.
Trans-isopropylN-1-3-(tert-butylsulfamoyl)-4-12-[4-
(isopropoxycarbonylamino)cyclohexyl]
thiazol-5-yliphenylicarbamate (Compound S12). NMR (400MHz, METHANOL-d4) 6 =
8.37 (d, J=2.3 Hz, 1H), 7.75 (s, 1H), 7.69 (dd, J=2.2, 8.4 Hz, 1H), 7.40 (d,
J=8.3 Hz, 1H),
5.01 (td, J=6.3, 12.5 Hz, 1H), 4.86 - 4.82 (m, 1H), 3.52 - 3.42 (m, 1H), 3.04
(tt, J=3.5, 12.0
Hz, 1H), 2.35 - 2.19 (m, 2H), 2.15 - 1.99 (m, 2H), 1.72 (dq, J=3.0, 12.9 Hz,
2H), 1.43 (dq,
J=3.3, 12.6 Hz, 2H), 1.34 (d, J=6.2 Hz, 6H), 1.25 (br d, J=6.1 Hz, 6H), 1.14
(s, 9H). ESI
[M+H] = 581.2.
Cis-isopropylN-1-3-(tert-butylsulfamoyl)-4-12-14-
(isopropoxycarbonylamino)cyclohexylithiazol-5-yliphenylicarbamate (Compound
S13). 1-H
NMR (400MHz, METHANOL-d4) 6 = 8.37 (d, J=2.2 Hz, 1H), 7.76 (s, 1H), 7.70 (dd,
J=2.2,
8.3 Hz, 1H), 7.40 (d, J=8.3 Hz, 1H), 5.01 (td, J=6.3, 12.5 Hz, 1H), 4.84 (br
s, 1H), 3.77 (br s,
1H), 3.23 - 3.15 (m, 1H), 2.05 - 1.98 (m, 4H), 1.89 - 1.72 (m, 4H), 1.34 (d,
J=6.2 Hz, 6H),
1.25 (d, J=6.1 Hz, 6H), 1.16 (s, 9H). ESI [M+H] = 581.2
Example 2. Compound Primary Screening
BACKGROUND
Primary screening was a phenotypic screen that utilized the synthetic lethal
interaction between AID and RAD51 to identify compounds that were both potent
and on
target. AID expressing cells are dependent upon RAD51 for survival; inhibiting
RAD51 in
AID positive cells results in a cytotoxic effect. Based on such an effect,
compounds that were
potent in AID positive cells and were signficiantly less potent in AID
negative cells were
identified.
M4TERIALS AND SUPPLIES
Plastic ware and consumables needed for this experiment include: Cell Culture
media;
Evaporation Buffer media; 100% DMSO; 96 well U-bottom sterile culture plates;
250mL
bottle; 1.5mL Opaque amber epi tubes; Epi Tube rack; 300mL reservoirs; 25mL
reservoir;
25mL serological pipette tips; 5mL serological pipette tips P1000 Pipette
Tips; and P200
Pipette Tips.

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
Equipment needed for this experiment include: Viaflo 384 liquid handler;
Eppendorf
serological pipette; Eppendorf P1000 Pipette; and Eppendorf P200 Pipette
Daudi Cell Culture and WI-38 Cell Cultures werealso needed for this
experiment.
Lastly, compounds (e.g., the compounds of this application) to be tested are
needed.
PROCEDURE
All steps were performed in a sterile environment inside the Biosafety
cabinet.
The first step was to set up a cell killing assay in the Daudi cell line (AID
positive). A
96 well u-bottom plate was prepared by writing the experiment number, plate
number, date
and initials in the top right corner of the plate lid. With a sterile 300m1
reservoir, and 25m1
serological pipette, evaporation buffer media was pipetted into reservoir in
25m1 increments.
Using the liquid handler,150u1 of evaporation buffer media was pipetted from
reservoir into
rows A and H, and Columns 1 and 12 of the 96 well u-bottom plate. Cell
cultures were
counted to obtain the density of cells per ml, and the culture viability. The
cell density
information was used to obtain 1,000,000 cells from culture using a 5mL
serological pipette
into an epi tube. The cell density information from the culture was used to
calculate the
number of cells and volume of media needed for the assay to seed 1250 cells in
130u1 of
media per available culture well in the 96 well u-bottom plate. Rows B through
F were used
for cells (50 wells in total), with row G left for an empty media control. The
calculation was
overestimated by I OmL to account for the dead volume in the 300m1 reservoir.
Once the
media volume was calculated, the appropriate volume of media was pipetted in
25mL
increments into the 250mL bottle using a 25mL serological pipette. The 250m1
bottle was
capped tightly, and placed into a 37 C water bath for 2 minutes. While the
culture media was
warming, I OmL of fresh media was pipetted from the 500mL culture media bottle
into a
sterile 25mL reservoir. Using the Eppendorf multichannel pipette, 130u1 of
media was piptted
from the 25mL reservoir into row G of the 96 well u-bottom plate. Once the
250mL bottle of
media was warmed, the volume of culture needed was pipetted into the bottle,
and mixed
gently with a 25mL serological pipette as to not create bubbles, and then the
contents of the
bottle were pipetted into a new 300mL reservoir. Using the liquid handler,
130u1 of culture
was pipetted from the 300mL reservoir into rows B through F of the 96 well u-
bottom plate.
Once the culture was added, the plate was placed into a 37 C incubator until
the compound
master plate was prepared for use.
Two 96 well u-bottom plates were prepared by writing the master plate name in
the
upper right corner of the plate lid. Labeling one DMSO master and the other
Media Master.
86

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
The compounds of interest were obtained from the laboratory freezer, and
placed into a 25
well storage box with a lid, and set the box aside. The compounds were
vortexed after
thawing but before use. Using an automatic multichannel pipette, 20u1 of 100%
DMSO was
pipetted into wells B3-B11 through G3-G11 of the DMSO master plate. For each
compound
on the master plate, 50u1 of the compound were pipetted in the appropriate
well of row 2
(reference plate map to determine appropriate well). A serial dilution was
prepared beginning
by aspirating 20u1 from row 2 and mixing with row 3, repeating until row 11
was reached.
Using the liquid handler, 194u1 of Daudi media was dispensed into wells B2-B11
through
G2-G11 of the Media master plate. Using the liquid handler, 6u1 from the DMSO
master
plate was aspirated and dispensed into the media master plate, mixing 100u1
twice.
Compounds from master plate were then added to the culture plate. The culture
plates
were removed from the incubator, and set inside the biosafety cabinet. Using a
liquid handler,
20u1 from wells B2 to B11 through G2 to Gil of master plate were aspirated,
and dispensed
into wells B2 to B11 through G2 to Gil of culture plate. This set was
continued with each
culture plate. Once the culture plates acquired their 20u1 of compound
dilutions, they were
placed back into the incubator, until their reads on Day 7 of experiment. Cell
death was
measured on Day 7 of the experiment using Cell-Titer Glo and a Promega Plate
reader.
Percent cell death and ECso values were calculated by comparing the cell
viability of
the compound treated wells to the non-treated wells. Normalized RLU values
were obtained
by subtracting the media well values from each of the wells in the same
column, and then
dividing that value by the DMSO treated cells values. The percent kill was
then calculated by
subtracting the normalized RLU value from 1 and multiplying by 100. The
average
normalized percent kill value and standard error of the mean was then
calculated. The kill
values were then inputted into Prism with the corresponding standard errors.
In Prism a non-
linear regression line was plotted with the data points using a semi-log
scale, and the ECso
value was calculated. For compounds that showed good potency in the Daudi cell
line, the
assay was repeated using WI-38 cells (AID negative).
87

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
Screening Data
Table 1. Compounds of the Present Application
AID+ EC50 AID- ECso
(111M) (111M)
A = < 0.1 julVI A = < 0.1 uIVI
Cmpd.
Structure B = < 1 tilVI B = < 1 tilVI
No.
C = > 1 tilVI C = > 1 tilVI
ND = Not ND = Not
Determined Determined
N 0)__ 1p
I ,___NO_ NH
0
1 0 & oS C ND
(rci)LN W µ J
H ,i, 'N
\--N 0H
\
0
Ill I SN,Nao)___NH
2 C ND
T'ss(21iN 0 ,P J
H l'i
\¨N
0,µ
N / N )_ >-NH
I A C 0
S \
3
0 110 I, k
110 ")L" OH
N
1 )----CN--e
ij34
N / S
4 6 o o--< C C
,
! N N .. S/,
1 H H cl ri
N
c)).LN 43 0
C ND
/So
H HN
)\
N
I i, >-N"' _-/
I 0 S \__I \\
0 0
C ND
6
0 0 ENI
HNISC)
)\
0 p
N
I )======0 Y-0
7 0 B ND
S
A
- 0 N 0
4
H "N
0 H
88

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
AID+ ECso AID- ECso
(AM) (11,M)
A = < 0.1 iitlVI A = < 0.1 iitlVI
Cmpd.
Structure B = < 1 tiM B = < 1 tiM
No.
C = > 1 tiM C = > 1 tiM
ND = Not ND = Not
Determined Determined
o
fzio
N
"'NH /
8
1 1 0 NI
os C ND
.,
0 N /S,
H o' N
NI
0A ,P)/
N
"'NH /
9 o B ND
j 0 os
N 4 ,<
H 4 N
0H
N
ilil
C?
0
)L , H A C
0 s / 0:
1-0
N 0
1 \
1 io s- ,,,N N
0 S)"....0 H)\--C(6
11 11,-0 / B ND
110 El Vi I
HNIK
N
/ .._.0 0 is),
12 )0, 0
0 s
,,,0 'NHi
/N
B ND
40 NH NH S'
HNI,...
, N 0
._F 13 0 (s) tio
N 0 N A C
N H
H
HL-
O N"---N
H
, N
0 AL %)---__
14 0 (R) NA N W / S --t N A C
H
õ..k-
oi N
H
N
F 0 ()
1 )
N = B o
(s) NN OS 'N A C
..-- O H
= H
N
0 / \ 0 \.._
16 411#
No j 11 osrsi,---.6/ ----
1 R Nr-NN A C
= H
89

CA 03133005 2021-09-08
WO 2020/198298 . PCT/US2020/024601
AID+ ECso AID- ECso
(11,M) (11,M)
A = < 0.1 M A = < 0.1 NI
Cmpd.
Structure B = < 1 pIVI B = < 1 tiM
No.
C = > 1 tiM C = > 1 tiM
ND = Not ND = Not
Determined Determined
0 )
17 s B C
0 0
s,
# N
H OH
0
N r
1 --)--NH
18 s
5...), /01 /5) k A C
40 " -.--N
O H
0,
N
I ,¨\'-NH
19 0 s
0 I = , A C
).L g/, )c
K'N---1-/-1 N # N
OH
11 1
`---.%
I c-NH
20 0 s C ND
o
ON 4 .<
H 4N
...., H
N
i ),....0c-NH
21 Ao 110 s A ND
o
1110 N N
S,
H H õi/ N
w H
0 \)---
N ,-0
1 )=-<---)NH
22 s B ND
9 0 0
)<)1-.. #
s,
#
0/-1 " I N
OH
N V
N
0 0 S)....."0õNe---o
23 -
40 NHINH .411113-VP. So A ND(
F 1,N11-1

CA 03133005 2021-09-08
WO 2020/198298 ' PCT/US2020/024601
AID+ ECso AID- ECso
(AM) (11IM)
A = < 0.1 uIVI A = < 0.1 iiiM
Cmpd.
Structure B = < 1 tilVI B = < 1 tiM
No.
C = > 1 tiM C = > 1 tiM
ND = Not ND = Not
Determined Determined
N 0 \
0 0 1 SOõ'N)LO
24
01 (R) hiAH 0 H A C
s'::
1 -o
N 0 \
/

N NIN 5 P H A ND
,C0
H H
HN<__
I -'
, N
/ \ 0
- 0 0
õ
N)\-0
26 it o H A C
0 (s) ii-- hi s---:.
--...._.,.NH
N
A C
27 F 0 N AN 0
S-:: H
H H I '0
N
\ 0
28
F 0 0 1 õ'N)Lo
o H A ND
0 Vi..-11, [1 s--
-:.
--..,.._,..NH
i N 0 )____.
0 0 ' S>\''.."0õ,N)\---0
29 0 H A C
jN"-",-------ILN S?::
I H H 'O
====...,õ ..,_,õNH
I NON/H
s ___
0 =P A C
k
s,
N,, (,) N'ILN ii N
H H OH
,
0
N yo
I ____________________________ ). "NH
S A C
31
01 lip i ,\<.
õ,õ,m,---,1 ei, ,ri
L.-.F
91

CA 03133005 2021-09-08
WO 2020/198298 . PCT/US2020/024601
AID+ ECso AID- ECso
(AM) (11IM)
A = < 0.1 juIVI A = < 0.1 iiiM
Cmpd.
Structure B = < 1 tiM B = < 1 tiM
No.
C = > 1 tiM C = > 1 tiM
ND = Not ND = Not
Determined Determined
0
N yo
I -)...NH
32 s A C
0 is p k
N ' N
1 --. (s) N H 01/S-FIN
N
0 HO--N1$/ s,...õ.\----/
., 09-....__\
33 0.-1(
- N 0 C ND
H
H
/ N\
010?)õ \ N IT =s)---0 )\---0
34 0¨NN=*0 B ND
H
H
0 N.---
H
0 / N0
C5s\OA th SrOi X
35 N [1 =/ s,0 '"N B ND
H
H
, N 0 \
0
36 (R?'"NcIAN W e B ND
H
\--NH H
0 N
H
N
s
37 (sNOIN 10 ,0 .'N)L()
H B ND
\--NH H /Si, ,IK
0/ N
H
N 0
/ SED )L 0
38 0a ), 0 ,
NO
0 H H A C
0 N SC
1 0
HN1.
'NH
39 0 s A C
0
4 x 40 0A1 1
s, H
I )---0 \---0
'NH
40 F 0 S A C
io A 0
4 x
0 iri
92

CA 03133005 2021-09-08
WO 2020/198298 . PCT/US2020/024601
AID+ ECso AID- ECso
(1LNI) (jiM)
Cmpd. A = < 0.1 julVI A = < 0.1 iuM
Structure B = < 1 tilVI B = < 1 tilVI
No.
C = > 1 tiM C = > 1 tiM
ND = Not ND = Not
Determined Determined
\---0
'NH
41 - o
, sA C
, It 0
1110 m CY 'N
H 4 N
w H
)---0
NH
42 0 s A C
o
,Asi,,,oN
I H ii N
0 H
)---
,---0
NH
43 0 s B C
)-L o
401 (R)
H
N 0y___
i s)......0
'NH ---0
"
44 0 1 C ND
A Op
-0 N iSi,
H 01 El
0 y
N
45 A ND
0 0
c.)...., N N 0 H # N
1 /, H
N0 _____.
1 "--N---N?-\-' -c)
46 1 )C.) SS /53C C
0 N
H 01 iNil
93

CA 03133005 2021-09-08
WO 2020/198298 . PCT/US2020/024601
AID+ ECso AID- ECso
(111M) (111M)
A = < 0.1 julVI A = < 0.1 uIVI
Cmpd.
Structure B = < 1 tilVI B = < 1 tilVI
No.
C = > 1 tilVI C = > 1 tilVI
ND = Not ND = Not
Determined Determined
, N
3
47 Ao fa 43s)."(1) \----o A C
110 11 Ill H
Ht.
N 0 I
1 '0 )LOro
48 0 s
)^ r P "'N
H B C
H
HN
, N 0 r
I
/ \
0 0 S).-'0õ,N)\----0
49 Oa A=p
H B C
0 N Si cc,
H
HN
N 0
/ >....0 0)\_.. r I
o
50 N , I ifil /OS "' N
H A C
; N N '. Si--
1 H H , -0
HN
(
0
N
51 , o . s ",NH A C
: A ,o
0 (s) N N S',
H H 6' EN,
_0.1
0
N
).....0 ,---0
52 o f 10 s ",NH A C
A ,o
(R) N N ,Si.
H H 01, ,N
_s)
0 __
N
53 F 9 a 1 pS).0 NH A C
*NNEiE ,s, ,<
0/ EN,
94

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
AID+ ECso AID- ECso
(AM) (jiM)
A=<0.1 julN4 A=<0.1 iiiM
Cmpd.
Structure B = < 1 tiM B = < 1 tiM
No.
C = > 1 tiM C = > 1 tiM
ND = Not ND = Not
Determined Determined
____13
o
N
I ,---0 \---0
54 F I a 43,S "'NH A C
40 (R) 11 11 ,S,
OH N
O
v-01
0
N
1 )....0 )--07¨
55 F = 0 [10 s
(s)N A C
A N
so
H H d N
N N=)_
1 s\>i / NH )¨
56 140 ,0 Me0 Oe¨ B ND
rolii
HNN
HN,..........-
)C
F
,
N} N Ca
/
9 Si ,ps)õ,N)L-0/ -
57 H A C
SI ENI , 0
HNS:-.
F
N 0
,1o)CD )L0)
AO S
58 H B C
0 N 'CO
H
HN
N
1 ....0
0 a)L0)--
59 9 io os
0j< H A C
0 N
H
N
0
I s)..--t 03
'"NH
59A o a 0 B C
A
0 [1 [1 'w ,s,
0/ N
0 _____
59B Ailo
N 110 I / A C
o
, ,<
dr ri

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
AID+ ECso AID- ECso
(AM) (11,M)
A=<0.1 julN4 A=<0.1 julN4
Cmpd.
Structure B = < 1 tiM B = < 1 tiM
No.
C = > 1 tiM C = > 1 tiM
ND = Not ND = Not
Determined Determined
N 0 0 _____
1 >=====0 ---0
NH
60 1 1 0
0 N #
:
s B C
H ,."N
N 0 0
60A C C
o
o)LN 4
H õ// N
/ H
N 0 0 _____
60B 0 I s (R) (S) N B C
o
S,
H õ/ N
/ H
N
/ 61 1110 isil=N 0 ).____,t;\____ 0 _
0
S 0,11
r C C
//
H H //0 1_,
S,
0H
N
N
62
i c lyNNic
9 _
-- H N * 0 0 H C C
H //
õs,
0 N---\
H
,---0
1 NH
63 0 B ND
4
(:))LN S,
H 0/ [1
N HN
I >----0 ---0
NH
64 )0L Fa s A C
o
[10 N N
H H /4 -.<
0/ 11
----0
I N>'''''<>-.NH
65 0 S B C
/0
OAN /Pi' N
H 0H
96

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
AID+ ECso AID- ECso
(AM) (11IM)
A=< 0.1 julN4 A=< 0.1 iiiM
Cmpd.
Structure B = < 1 tiM B = < 1 tiM
No.
C = > 1 tiM C = > 1 tiM
ND = Not ND = Not
Determined Determined
o / Ni)L0 9, \
/00 NA * S "---cc"--
'"N
66A H N
H ,
S( H A C
i NH-o
.....)..-
41 HNJZHN 40 c:0S)N '''.0',N3L)
66B ---
H B ND
r*o
_...)...-NH
(N\
67A H
0 ith / 0 IDAN
s,0
'N
H B C
/ ,)o `,....NH
N
67B N
cr. j0( *
H ,0
H C ND
sZ
/ `0
...),NH
N 0
I 2-No____ y-o
68 o a i NH
B ND
)L
NH ri /4
0/ N
N
I 69 oJ(I %___ )---0
s/ NO_N H
C ND
0
cps)LN 4
H "N
I NH
70 F 0 A C
o
10/ ON 4
# N
0 H
I ,---0 >---0
'NH
71A C
10 (s) o N 4
H õC/ N
k., H
97

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
AID+ ECso AID- ECso
(AM) (11IM)
A = < 0.1 u1VI A = < 0.1 u1VI
Cmpd.
Structure B = < 1 p1VI B = < 1 p1VI
No.
C = > 1 p1VI C = > 1 p1VI
ND = Not ND = Not
Determined Determined
I s>----"O """ NH
72 o A C
o
4 ,<
I H /I N
0 H
73 o
os A C
6 NN 4 ,<
H H cr N
HO
74 o A C
)-L p
0 N
N ill /SI, X0H
H
01
H
Example 3. Bi-directional Caco-2 permeability
Bi-directional Caco-2 permeability was assayed. Caco-2 cells were seeded onto
permeable polycarbonate supports and allowed to differentiate for about 3
weeks prior to
being used in the assays. The cells were then exposed to the compounds from
either the
apical or basolateral sides and incubated at 37C for up to 90 minutes under
light agitation.
Compound transport was then measured using LC/MS/MS analysis at 30, 60, and 90
minutes.
Table 2
Caco-2 Results
Compound
AB Papp BA Papp BA/AB AB BA
No.
(cm/sec x 106) (cm/sec x 106) Ratio Recovery % Recovery %
5.9 2.2 0.4 34.7 80.8
7 1.7 2.1 1.2 55.9 92
1.2 1.5 1.3 59.9 87.2
17 2.8 2.1 0.8 47.2 81.9
9.4 10.4 1.1 44.8 96.6
21 3.6 10.7 3 58.6 78.3
98

CA 03133005 2021-09-08
WO 2020/198298
PCT/US2020/024601
Caco-2 Results
Compound
No. AB Papp BA Papp BA/AB AB BA
(cm/sec x 106) (cm/sec x 106) Ratio Recovery % Recovery %
1.1 21 18.7 83.1 89
22
0.7 0.5 0.7 92 107.1
27
1.9 3.3 1.7 66.9 81
28
10.9 24.1 2.2 93.9 90.9
29
11.5 15.7 1.4 79.8 115.6
12.8 13.6 1.1 70.5 92.3
31
0.4 45.2 103.9 98.2 96.8
34
0.4 41 99.1 98.8 107.5
17.9 22.9 1.3 73.3 82.3
38
3.2 4.2 1.3 36.2 69.4
44
7.6 10.8 1.4 73.6 84.4
8.7 12.4 1.4 65.3 80.7
47
23.1 16 0.7 80.1 90.3
48
2 29.1 14.6 87.7 98.1
9.9 10.5 1.1 65 85.7
51
7.4 10.9 1.5 64.1 91.7
52
9.3 9.7 1 63.1 90.9
53
6.5 7 1.1 65.2 83.8
1.3 3.5 2.7 61.9 86.8
57
5.1 3.4 0.7 52.3 83.5
58
3.1 15 4.8 61.5 91.5
59
3.1 9.6 3.1 55.6 85.5
59A
4.6 9.7 2.1 56.2 82
59B
15.6 13.3 0.9 59.8 90.8
99

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
Caco-2 Results
Compound
AB Papp BA Papp BA/AB AB BA
No.
(cm/sec x 106) (cm/sec x 106) Ratio Recovery % Recovery %
60A 12 11.2 0.9 55.3 86
60B 11.6 13.5 1.2 51.2 84.3
63 10.5 26.7 2.5 83.6 90
64 2.2 27.4 12.5 79.7 95.9
66A 2.9 3.9 1.4 64.9 81.2
67A 10.5 6.9 0.7 50.7 78.6
Example 4. Human Liver Microsome Stability
The stability of the claimed compounds was determined in the presences of
human
liver microsomes. The compounds were incubated with the microsomes at 37 C
for 45
minutes. Samples were analyzed using LC-MS/MS. Data analysis included half-
life,
clearance rate, and the percentage of hepatic blood flow (%QH) for each of the
compounds in
the different species. Below are liver microsome assat data of representative
compounds,
which show that the claimed compounds have superior metabolic stability.
Table 3
Compound Human Liver Microsome Stability
No. Half Life (min)
Clearance (m/min/mg) % QH
29 20.5 68.0 78.8
31 22.4 61.9 77.3
66A 77.6 18.2 49.6
67A >300 <4.6 <20.3
Example 5. Cell Line Screen
The activity of the claimed compounds was measured in a variety of cell lines
with
different expression levels of activation induced cytidine deaminase (AICDA).
The potency
assay was repeated in all of the listed cell lines and the ECso values
recorded.
Table 4
EC50 (nM) EC50 (nM)
01M)
Cell Line (Cancer AICDA EC50 (nM) EC50 Cmpd. Cmpd.
Cmpd. 29 Cmpd. 31
Type) Expression 66A 67A
Daudi (Lymphoma) High 43 20 18 311
WSU-FSCCL
Negative 67 <40 25 344
(Lymphoma)
100

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
EC50 (nM) EC50 (nM)
EC50 (nM) EC50 (nM)
Cell Line (Cancer AICDA
Cmpd. 29 Cmpd. 31 Cmpd. Cmpd.
Type) Expression 66A 67A
U-698-M
High 113 31 88 791
(Lymphoma)
CCRF-SB
High 1283 2164 183 932
(Leukemia)
KYSE-70
Low 4660 4701 2639 2629
(Head and Neck)
SNU-1
Negative n.d. n.d. 609 2927
(Gastric)
KG-1
Negative >10000 8785 3067 2995
(Leukemia)
KYSE-510
Negative >10000 >10000 4516 3403
(Head and Neck)
SNU-5
Low n.d. n.d. 2941 3845
(Gastric)
TOV-1120D
Negative 9172 n.d. 2377 4924
(Ovary)
OV56
Low 9086 n.d. 5944 7228
(Ovary)
ARPE19/HPV16
(HPV Immortalized Negative >10000 >10000 >10000 >10000
RPE)
WI-38 (Normal
Human Lung Negative >10000 >10000 >10000 >10000
Fibroblast)
n.d. not determined
Example 6. Pharmacokinetic (PK)
PK studies in mice were used to determine the fate of the compounds in a whole

organism. Rats were treated with the compounds either orally of via IV at the
indicated doses
and followed for up to 24 hours. Plasma samples were taken at different time
points and
analyzed by LC-MS.
Table 5
po @ 80 mg/kg (Formulation: 30%
Cmpd. Cmpd.
PEG400, 10% Vitamin E TPGS in Cmpd. 29 Cmpd. 31
66A 67A
water)
T112 (hr) 4.66 4.75 2.59 11.5
Rat Female
F (%) @5 mg/kg 8.39 2.77 3.31 86.5
T112 (hr) 3.97 3.79 1.86 6.46
Rat Male
F (%) @5 mg/kg 3.69 1.49 2.55 46.9
Example 7. Combination treatment of Compound 67A with PARP inhibitors
Genomic instability is a driver of tumorigenesis and cancer progression. Loss
of
tumor suppressors or activation of oncogenes can induce DNA damage stress,
promoting
genomic instability and creating dependencies upon key DNA repair pathways.
These
dependencies can be targeted therapeutically to induce synthetic lethality.
Without being
101

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
bound by any theory, RAD51 inhibitor, Compound 67A, which is selectively
active in
Activation Induced Cytidine Deaminase (AID) expressing cells. In cancer cells,
AID causes
significant genotoxic stress through DNA replication fork collapse which
creates a
dependency upon the homologous recombination repair factor, RAD51, for
survival.
Compound 67A acts by destabilizing RAD51 focus formation, leading to it
premature
nuclear export and subsequent degradation. PARP inhibitors use another
synthetic lethal
mechanism, in which PARP1, a protein important for repairing single strand
breaks, is
inhibited in BRCA1/2 deficient cancers. A main resistance mechanism to PARP
inhibitors is
the overexpression of RAD51; therefore, Compound 67A could act as a sensitizer
to PARP
inhibitors.
A matrix study was performed with Compound 67A (concentration range of 20nM to

M) and 5 different PARP inhibitors including olaparib (20nM to 2.5 M),
niraparib (20nM
to 2.5 M), veliparib (2.5 M to 50[tM), rucaparib (156nm to 10[tM), and
talazoparib (9nM to
0.625[tM). The combination matrix was tested in 3 cell lines of varying AID
expression:
ARPE19/HPV16 (HPV immortalized normal epithelial cell line), KYSE-70 (head and
neck
cancer cell line) and Daudi (Burkitt's Lymphoma cell line). PARP indicated
cells lines were
also tested including HCC1143 and BT20, both of which are derived from triple
negative
breast cancers and were selected for their varying responsiveness to olaparib.
Both the Loewe
Additivity model and the Bliss Independence model were used to determine drug
interaction
(synergistic, independent, or antagonistic). In general, synergy with the PARP
inhibitors was
observed in the tumor derived cell lines. Only with olaparib was synergy
observed in the non-
tumor derived cell line ARPE19/HPV16. Greater synergistic activity with
Compound 67A
with increasing PARP trapping efficiency was observed. Veliparib, as a pure
catalytic
inhibitor of PARP, deviated from that trend and showed similar levels of
synergy as that
observed with niraparib. These data suggest that Compound 67A may be active as
a
combinatorial therapy with PARP inhibitors. This Example shows there is
significant synergy
in combining RAD51 and PARP inhibition as a cancer treatment strategy.
Bliss Independence model was used and calculated in Excel. The model assumes
that
the drugs act with probabilistic independence.
Bliss: v
exp = YA YB YAYB
This expected value was generated by examining single-agent effects and then
tested
against the observed value using a two-tailed Welch's T-Test. For
visualization purposes, the
102

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
values generated from this test were transformed to a "SynergyScore" by doing
the
computation:
¨log(p) t
Synergy Score ¨ log(0.05)X iti
Loewe Additivity model was used and calculated by the "Synergyfinder" R
package.
X2
______________________________________________ =1
f1-1(YLoewe) f2-1(YLoewe)
The graphs generated describe the difference between the expected value
generated by the
additivity model and the observed value generated by the experiment. If the
effect determined
by the additivity model < the effect observed the space is colored red, if the
effect determined
by the additivity mode > the effect observed then the space is colored green.
The saturation of
the color is proportional to the magnitude of the difference between these two
values.
Table 6. Results from Combination Compound 67A + PARP Inhibitor Treatment in
Cells
Drug/Combination Cell Conditions Tested Conclusion
All cells tested showed strong
Compound VA AICDA Expression Range
synergy with olaparib regardless of
/olaparib (Low, Med, High)
AICDA expression
In both conditions combination
Compound 67A
BRCA-Status (wt, -/-) treatment showed concentration-
/olaparib
dependent synergy and antagonism.
The combination was strongly
Compound 67A AICDA Expression Range
/veliparib (Low, Med, High) synergistic in AICDA-low and
AICDA-high cells.
In both conditions combination
Compound 67A
BRCA-Status (wt, -/-) treatment showed concentration-
/veliparib
dependent synergy and antagonism.
The combination was synergistic
Compound 67A AICDA Expression Range
only in the AICDA-high cells at
/rucaparib (Low, Med, High)
low concentrations
The combination was synergistic in
Compound 67A AICDA Expression Range
AICDA-high cells and in AICDA
/talazoparib (Low, Med, High)
low cells.
The combination was strongly
Compound 67A AICDA Expression Range
/niraparib (Low, Med, High) synergistic in AICDA-low and
AICDA-high cells.
103

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
Example 8. Treatment with Compound 67A in Combination with Olaparib
Immunodeficient mice were subcutaneously engrafted in the rear flank with the
tumor
cells. The tumors were allowed to grow to a size of about 100 to about 200mm3
prior to being
randomized and grouped into treatment arms. For models HBCx11 through HBCx1
the four
treatment arms were vehicle, Compound 67A at 80 mg/kg QD, olaparib at 100
mg/kg QD,
and Compound 67A + Olaparib at 80 mg/kg QD and 100 mg/kg QD, respectively. For

models J000101173 through TM00091 the treatment arms were vehicle, Compound
67A at
40 mg/kg QD, olaparib at 50 mg/kg QD, and Compound 67A + Olaparib at 40 mg/kg
QD
and 50 mg/kg QD, respectively. The tumors were measured either once weekly or
twice
weekly.
Percent Tumor Growth Inhibition (%TGI) was calculated by taking the mean
volume
of each of the treatment arms, and comparing the treatment arms to the vehicle
arms using the
following equation:
(-treatment arm mean volume
___________________________________________ *
100 - TGIcY=
vehicle mean volume )
Results for each treatment arm are present in Table 7.
Table 7. Cellular Growth Inhibition by the Combination of Compound 67A and
Olaparib
Model % TGI
HRD RAD51 BRCA Status PALB2
67A Olaparib Combo Status Exp. Status
HBCx11 23.39 60.95 88.42 Positive High
BRCA1 Mut WT
HBCx17 -51.13 122 124.2 Positive High
BRCA2 Mut WT
HBCx23 NA 84.48 78.79 Positive High
BRCA1 Meth WT
HBCx27 13.14 7.819 51.50 Negative High WT WT
T298 NA 107 101 Positive Low WT Mut
HBCx16 -22.45 -25.61 -22 Positive High
BRCA1 Meth WT
HBCx12B -3.27 42.56 57.26 Positive High
BRCA1 Meth WT
HBCx1 28.75 51.07 114.9 Positive High WT WT
J000101173 19.87 4.71 -27.7 Positive Low WT WT
J000100675 36.69 -17 48.07 Negative Low WT WT
TM000113 7.24 2.37 45.74 Negative Low WT WT
TM01079 26.73 68.7 99.69 Positive Low
BRCA1 Meth WT
TM00090 51.82 86.05 109.8 Positive Low
BRCA1 Meth; WT
BRCA2 Mut
TM00096 -25.4 -1.94 -33.2 Positive Low WT WT
TM00091 27 21.26 61.79 Positive High
BRCA1 Mut WT
104

CA 03133005 2021-09-08
WO 2020/198298 PCT/US2020/024601
EQUIVALENTS
The details of one or more embodiments of the invention are set forth in the
accompanying description above. Although any methods and materials similar or
equivalent to
those described herein can be used in the practice or testing of the present
disclosure, the
preferred methods and materials are now described. Other features, objects,
and advantages of
the disclosure will be apparent from the description and from the claims. In
the specification
and the appended claims, the singular forms include plural referents unless
the context clearly
dictates otherwise. Unless defined otherwise, all technical and scientific
terms used herein have
the same meaning as commonly understood by one of ordinary skill in the art to
which this
disclosure belongs. All patents and publications cited in this specification
are incorporated by
reference.
The foregoing description has been presented only for the purposes of
illustration and
is not intended to limit the invention to the precise form disclosed, but by
the claims appended
hereto.
105

Representative Drawing
A single figure which represents the drawing illustrating the invention.
Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2020-03-25
(87) PCT Publication Date 2020-10-01
(85) National Entry 2021-09-08
Examination Requested 2022-09-29

Abandonment History

There is no abandonment history.

Maintenance Fee

Last Payment of $100.00 was received on 2022-12-13


 Upcoming maintenance fee amounts

Description Date Amount
Next Payment if small entity fee 2024-03-25 $50.00
Next Payment if standard fee 2024-03-25 $125.00

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee 2021-09-08 $408.00 2021-09-08
Maintenance Fee - Application - New Act 2 2022-03-25 $100.00 2022-02-22
Request for Examination 2024-03-25 $814.37 2022-09-29
Maintenance Fee - Application - New Act 3 2023-03-27 $100.00 2022-12-13
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
CYTEIR THERAPEUTICS, INC.
Past Owners on Record
None
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Abstract 2021-09-08 2 75
Claims 2021-09-08 12 454
Drawings 2021-09-08 45 3,793
Description 2021-09-08 105 5,242
Representative Drawing 2021-09-08 1 12
Patent Cooperation Treaty (PCT) 2021-09-08 1 39
International Search Report 2021-09-08 2 57
Declaration 2021-09-08 1 22
National Entry Request 2021-09-08 6 209
Cover Page 2021-11-24 2 44
Request for Examination 2022-09-29 4 180
Examiner Requisition 2024-04-04 5 245